Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Virol ; 93(13)2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30996091

RESUMEN

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.


Asunto(s)
Infecciones por Adenoviridae/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Adenovirus Humanos/fisiología , ADN Helicasas/metabolismo , Replicación del ADN , Replicación Viral , Células A549 , Infecciones por Adenoviridae/virología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Ubiquitina/metabolismo
2.
Hum Mol Genet ; 25(9): 1836-45, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26945007

RESUMEN

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.


Asunto(s)
Carcinoma Medular/congénito , Receptor beta de Estrógeno/genética , Regulación Neoplásica de la Expresión Génica , Mutación de Línea Germinal/genética , Neoplasia Endocrina Múltiple Tipo 2a/genética , Neoplasia Endocrina Múltiple Tipo 2a/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Adulto , Carcinoma Medular/genética , Carcinoma Medular/metabolismo , Carcinoma Medular/patología , Proliferación Celular , Susceptibilidad a Enfermedades , Genotipo , Humanos , Masculino , Neoplasia Endocrina Múltiple Tipo 2a/patología , Linaje , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Regulación hacia Arriba , Adulto Joven
3.
Mol Carcinog ; 55(1): 15-26, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25408419

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Proteínas de la Membrana/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Invasividad Neoplásica , Pronóstico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proto-Oncogenes Mas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ensayo de Tumor de Célula Madre , Ubiquitinación
4.
Clin Cancer Res ; 30(7): 1352-1366, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-37921808

RESUMEN

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.


Asunto(s)
Simportadores , Neoplasias de la Tiroides , Ratones , Animales , Humanos , Vorinostat/farmacología , Pertecnetato de Sodio Tc 99m/metabolismo , Radioisótopos de Yodo/uso terapéutico , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/genética , Simportadores/genética , Simportadores/metabolismo , Inhibidores de Histona Desacetilasas , Línea Celular Tumoral
5.
Front Endocrinol (Lausanne) ; 13: 1061555, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36686426

RESUMEN

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.


Asunto(s)
Hipertiroidismo , Neoplasias de la Tiroides , Humanos , Antitiroideos/uso terapéutico , Radioisótopos de Yodo/uso terapéutico , Litio/uso terapéutico , Prednisona/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/radioterapia , Hipertiroidismo/radioterapia , Hipertiroidismo/tratamiento farmacológico , Resultado del Tratamiento
6.
Cell Chem Biol ; 29(3): 502-516.e7, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-34520744

RESUMEN

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.


Asunto(s)
Neoplasias , Simportadores , Transporte Biológico , Humanos , Simportadores/genética , Simportadores/metabolismo
7.
J Clin Endocrinol Metab ; 107(5): 1392-1406, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-34791326

RESUMEN

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.


Asunto(s)
MicroARNs , Neoplasias de la Tiroides , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Pronóstico , ARN Mensajero/genética , Factores de Riesgo , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología
8.
Neurobiol Dis ; 41(2): 421-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20971191

RESUMEN

Axon regeneration in the adult central nervous system (CNS) is prevented by inhibitory molecules present in myelin, which bind to a receptor complex that leads to downstream RhoGTP activation and axon growth cone collapse. Here, we compared expression of Citron kinase (Citron-K), a target molecule of RhoGTP in non-regenerating dorsal root ganglion neurons (DRGN) after dorsal column (DC) injury, and in regenerating DRGN after either sciatic nerve (SN) injury or preconditioning SN+DC lesion models. We show by microarray that Citron-K mRNA levels in DRGN of a non-regenerating DC injury model were elevated 2-fold compared to those of intact control DRGN. Conversely, Citron-K levels were reduced by 2 and 2.4-fold at 10 days post lesion in the regenerating SN and preconditioning SN+DC lesion models, respectively, compared to levels in control intact DRGN. Western blotting and immunohistochemistry confirmed these observations and localised Citron-K immunostaining to both DRGN and satellite glia. In dissociated, adult rat DRG cell cultures, 80% knockdown of Citron-K, in the presence of inhibitory concentrations of CNS myelin extract (CME), promoted significant disinhibited DRGN neurite outgrowth, only when cells were stimulated with neurotrophic factors. The levels of RhoGTP remained unchanged after Citron-K knockdown in the presence of CME while enhanced cofilin levels correlated with disinhibited DRGN neurite outgrowth. This observation suggests that Citron-K plays a role in axon growth downstream of Rho activation. We conclude that Citron-K regulates actin polymerisation downstream of RhoA and may offer a potentially novel therapeutic approach for promoting CNS axon regeneration.


Asunto(s)
Axones/enzimología , Cofilina 1/metabolismo , Conos de Crecimiento/enzimología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Regeneración Nerviosa/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Axones/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/fisiología , Masculino , Regeneración Nerviosa/genética , Polimerizacion , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Quinasas Asociadas a rho/fisiología
9.
J Cell Sci ; 122(Pt 18): 3393-402, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19706688

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Proteínas Represoras/metabolismo , Simportadores/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Animales , Antígenos CD/metabolismo , Caveolinas/metabolismo , Línea Celular , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular , Yoduros/metabolismo , Proteínas de la Membrana/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Transporte de Proteínas , Proto-Oncogenes Mas , Ratas , Fracciones Subcelulares/metabolismo , Tetraspanina 30
10.
Neurobiol Dis ; 39(3): 292-300, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20451608

RESUMEN

To test the possibility that phosphorylated epidermal growth factor receptor (pEGFR) mediates axon growth inhibition, we determined if pEGFR levels were raised in dorsal root ganglia (DRG) after non-regenerating dorsal column (DC) lesions and suppressed in regenerating sciatic nerve (SN) and preconditioning (P) SN+DC lesioned DRG. Levels of EGFR mRNA and protein in DRG were unchanged between control and all injury models. Satellite glia and not DRG neurons (DRGN) constitutively contained pEGFR and, only in PSN+DC rats, were levels significantly reduced in these cells. In vitro, siRNA mediated knockdown of EGFR (siEGFR) mRNA and protein was associated with suppressed RhoA activation, but fibroblast growth factor-2 (FGF2) was a mandatory requirement for DRGN neuritogenesis after addition of inhibitory concentrations of CNS myelin. Thus, EGFR activation in satellite glia was not consistently correlated with DRGN axogenesis and siEGFR reduction of pEGFR with attenuated Rho-GTP signalling did not promote DRGN disinhibited neurite outgrowth without exogenous FGF2 stimulation. Together, these data argue against a direct intra-axonal involvement of pEGFR in axon regeneration.


Asunto(s)
Axones/metabolismo , Receptores ErbB/metabolismo , Ganglios Espinales/metabolismo , Regeneración Nerviosa/fisiología , Neuroglía/metabolismo , Neuronas/metabolismo , Nervio Ciático/fisiología , Factor de Transcripción Activador 3/metabolismo , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Proteína GAP-43/metabolismo , Galanina/metabolismo , Inmunohistoquímica , Masculino , Compresión Nerviosa , Neuronas/citología , Neuropéptido Y/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Mol Cell Neurosci ; 40(4): 451-62, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19340932

RESUMEN

In investigating the consequences of gene silencing in axon growth disinhibition strategies in cultured retinal ganglion cells (RGC), we conducted experiments designed to silence RhoA signalling in PC12 and primary adult rat retinal cell cultures (containing RGC) by siRNA-mediated RhoA mRNA knockdown. We demonstrate wide differences in the levels of RhoA mRNA knockdown, dose-dependent cell toxicity, and induction of endogenous inflammatory cytokine and interferon responses to siRNA therapy. Toxicity effects observed with RhoA-siRNA was significantly reduced with "Stealth" chemical modification of the sequence, promoting approximately 50% and 70% knockdown of RhoA mRNA and protein in retinal cells, respectively, while promoting significant disinhibited RGC neurite outgrowth in the presence of inhibitory CNS myelin. Our results highlight differential responsiveness of cell lines compared to primary cultured cells, and demonstrate the efficacy of the "Stealth" modification to reduce siRNA-induced interferon responses, thereby increasing target cell viability and reducing off-target effects of the delivered nucleic acids.


Asunto(s)
Silenciador del Gen , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Células Ganglionares de la Retina/fisiología , Proteína de Unión al GTP rhoA , Animales , Células Cultivadas , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Citocinas/metabolismo , Técnicas de Silenciamiento del Gen , Interferones/metabolismo , Neuritas/metabolismo , Células PC12 , ARN Interferente Pequeño/genética , Ratas , Células Ganglionares de la Retina/citología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
12.
Cancer Res ; 80(1): 102-115, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672844

RESUMEN

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.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Aparato de Golgi/metabolismo , Radioisótopos de Yodo/farmacología , Simportadores/metabolismo , Cáncer Papilar Tiroideo/terapia , Neoplasias de la Tiroides/terapia , Proteína que Contiene Valosina/metabolismo , Adulto , Animales , Mama/patología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Membrana Celular/metabolismo , Quimioradioterapia/métodos , Femenino , Perfilación de la Expresión Génica , Humanos , Radioisótopos de Yodo/uso terapéutico , Estimación de Kaplan-Meier , Masculino , Ratones , Persona de Mediana Edad , Cultivo Primario de Células , Pronóstico , Supervivencia sin Progresión , Proteolisis , Cáncer Papilar Tiroideo/mortalidad , Cáncer Papilar Tiroideo/patología , Glándula Tiroides/citología , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/patología , Glándula Tiroides/efectos de la radiación , Neoplasias de la Tiroides/mortalidad , Neoplasias de la Tiroides/patología , Distribución Tisular , Proteína que Contiene Valosina/antagonistas & inhibidores
13.
J Gene Med ; 11(6): 523-34, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19322910

RESUMEN

BACKGROUND: A promising strategy to enhance axon regeneration is to employ short interfering (si)RNA targeting either RhoA or p75(NTR), which are components of a signalling cascade triggered by growth inhibitory ligands. However, it is important to profile the biological impact of siRNA on cell homeostasis in order to develop safe and effective therapies. METHODS: We used microarray and quantitative reverse transcriptase-polymerase chain reaction techniques to analyse the transcriptional effects of siRNA against p75(NTR) and RhoA in neuronal cell line and primary cultures. RESULTS: Expression analysis showed that primary rat dorsal root ganglion cells were up to 279-fold more sensitive than nerve growth factor-differentiated PC12 cells in detecting innate immune responses to siRNA. The sequence and method of synthesis of siRNA critically influenced the magnitude of the innate immune response. Importantly, siRNA sequences were identified that efficiently silenced RhoA and p75(NTR) mRNA with attenuated induction of the interferon-responsive gene mx1. Moreover, microarray analysis identified genes related to RhoA function, such as tgf beta 2, plod2 and mmp3, with implications for interpretating the ability of RhoA siRNA to promote axon regeneration. CONCLUSIONS: These findings demonstrate the importance of screening the biological impact of different siRNA sequences not only for their silencing efficacy, but also for potential toxicity. The results of the present study suggest that the toxicity observed was sequence-dependent because only two out of five siRNA sequences targeting RhoA were identified that did not induce a significant innate immune response.


Asunto(s)
Neuronas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/química , Animales , Células Cultivadas , Ganglios Espinales/metabolismo , Silenciador del Gen , Vectores Genéticos/genética , Inmunidad Innata , Neurogénesis , Células PC12 , Ratas , Receptor de Factor de Crecimiento Nervioso/genética , Transfección , Proteína de Unión al GTP rhoA/genética
14.
J Gene Med ; 11(4): 335-44, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19248141

RESUMEN

BACKGROUND: Polymer coating of adenovirus type 5 (Ad5) particles produces a 'stealth' Ad5 (sAd5) that confers protection from immune recognition, blocks receptor-mediated uptake, and favours uptake into pinocytic cells. METHODS: In mixed cultures of primary adult rat dorsal root ganglion neurones (DRGN), rat C6 glioma cells, A9 non-Coxsackie and Ad Receptor (CAR)- and CAR-expressing fibroblasts, reporter gene expression after sAd5 pinocytotic uptake was monitored using the green fluorescent protein (gfp) gene, and viral particle trafficking and polymer coat dismantling was followed using Yoyo-1 tagged Ad5 DNA and Texas Red (TR) to label the coat. RESULTS: sAd5.gfp was pinocytosed by significantly higher proportions of neurones, than other cells, but GFP was not expressed. The TR-labelled coat remained co-localised with tagged viral DNA within transfected DRGN, showing that sAd5 did not uncoat and viral DNA did not traffic to the nucleus. Noncoated Ad5 transduced non-neuronal DRG cells more efficiently than DRGN, whereas A9(CAR) cells were more significantly transduced than any other cell type. Retargeting of the sAd5.gfp with either fibroblast growth factor-2 or nerve growth factor (NGF) enhanced internalisation by DRGN into endocytic vesicles allowing uncoating and thus GFP expression. Retargeting with NGF resulted in significantly higher numbers of DRGN expressing GFP than non-neuronal DRG cells. CONCLUSIONS: These findings indicate that DRGN pinocytose atropic genetic particles at higher levels than non-neuronal DRG cells and the environment of pinocytic vesicles is not conducive to sAd5 uncoating and capsid dismantling, requiring reformulation of sAd5 with either a neurone specific ligand or a self-dismantling coat to target sAd5 transgene expression to neurones.


Asunto(s)
Adenoviridae/genética , Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Vectores Genéticos , Neuronas/metabolismo , Animales , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/metabolismo , Ganglios Espinales/citología , Glioma/metabolismo , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/genética , Factor de Crecimiento Nervioso/farmacología , Ratas
15.
Mol Cell Neurosci ; 39(3): 411-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18722531

RESUMEN

Adenovirus (Ad) is an efficient and safe vector for CNS gene delivery since it infects non-replicating neurons and does not cause insertional mutagenesis of host cell genomes. However, the promiscuous Ad CAR receptor targets cells non-specifically and activates a host immune response. Using Ad5 containing an expression cassette encoding the gene for green fluorescent protein, gfp, regulated by the neuron specific promoter synapsin-1 and the woodchuck post-transcriptional regulatory element (WPRE), we demonstrate efficient, prolonged and promoter-restricted gfp expression in neurons of mixed primary adult rat dorsal root ganglion (DRG) and retinal cell cultures. We also demonstrate restricted gfp expression in DRG neurons after direct injections of Ad5 containing the synapsin-1(gfp)/WPRE construct into L4 DRG in vivo, while Ad5 CMV(gfp) transfected both DRG glia and neurons. Moreover, since the effective titres of delivered Ad5 are reduced with this neuron specific promoter/WPRE expression cassette, the viral immune challenge should be attenuated when used in vivo.


Asunto(s)
Adenoviridae , Neuronas/fisiología , Neuronas/virología , Transgenes , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Células Cultivadas , Ganglios Espinales/citología , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Neuronas/citología , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Sinapsinas/genética , Sinapsinas/metabolismo
16.
Thyroid ; 29(10): 1485-1498, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31310151

RESUMEN

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.


Asunto(s)
Radioisótopos de Yodo/metabolismo , Multimerización de Proteína , Simportadores/metabolismo , Neoplasias de la Tiroides/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Transferencia Resonante de Energía de Fluorescencia , Células HeLa , Humanos , Inmunoprecipitación , Técnicas In Vitro , Conformación Proteica , Estructura Cuaternaria de Proteína , Simportadores/ultraestructura , Neoplasias de la Tiroides/radioterapia
17.
Sci Rep ; 9(1): 5120, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30914656

RESUMEN

Loss of latexin (LXN) expression negatively correlates with the prognosis of several human cancers. Despite association with numerous processes including haematopoietic stem cell (HSC) fate, inflammation and tumour suppression, a clearly defined biological role for LXN is still lacking. Therefore, we sought to understand LXN expression and function in the normal and malignant prostate to assess its potential as a therapeutic target. Our data demonstrate that LXN is highly expressed in normal prostate luminal cells but downregulated in high Gleason grade cancers. LXN protein is both cytosolic and secreted by prostate cells and expression is directly and potently upregulated by all-trans retinoic acid (atRA). Whilst overexpression of LXN in prostate epithelial basal cells did not affect cell fate, LXN overexpression in the luminal cancer line LNCaP reduced plating efficiency. Transcriptome analysis revealed that LXN overexpression had no direct effects on gene expression but had significant indirect effects on important genes involved in both retinoid metabolism and IFN-associated inflammatory responses. These data highlight a potential role for LXN in retinoid signaling and inflammatory pathways. Investigating the effects of LXN on immune cell function in the tumour microenvironment (TME) may reveal how observed intratumoural loss of LXN affects the prognosis of many adenocarcinomas.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Células PC-3 , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Supresoras de Tumor/genética
18.
Nucleic Acids Res ; 34(11): e80, 2006 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-16835307

RESUMEN

Non-viral vectors are promising vehicles for gene therapy but delivery of plasmid DNA to post-mitotic cells is challenging as nuclear entry is particularly inefficient. We have developed and evaluated a hybrid mRNA/DNA system designed to bypass the nuclear barrier to transfection and facilitate cytoplasmic gene expression. This system, based on co-delivery of mRNA(A64) encoding for T7 RNA polymerase (T7 RNAP) with a T7-driven plasmid, produced between 10- and 2200-fold higher gene expression in primary dorsal root ganglion neuronal (DRGN) cultures isolated from Sprague-Dawley rats compared to a cytomegalovirus (CMV)-driven plasmid, and 30-fold greater expression than the enhanced T7-based autogene plasmid pR011. Cell-free assays and in vitro transfections highlighted the versatility of this system with small quantities of T7 RNAP mRNA required to mediate expression at levels that were significantly greater than with the T7-driven plasmid alone or supplemented with T7 RNAP protein. We have also characterized a number of parameters, such as mRNA structure, intracellular stability and persistence of each nucleic acid component that represent important factors in determining the transfection efficiency of this hybrid expression system. The results from this study demonstrate that co-delivery of mRNA is a promising strategy to yield increased expression with plasmid DNA, and represents an important step towards improving the capability of non-viral vectors to mediate efficient gene transfer in cell types, such as in DRGN, where the nuclear membrane is a significant barrier to transfection.


Asunto(s)
Citoplasma/genética , ARN Polimerasas Dirigidas por ADN/genética , Regulación de la Expresión Génica , Neuronas/metabolismo , ARN Mensajero/metabolismo , Transfección/métodos , Proteínas Virales/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Citoplasma/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Expresión Génica , Humanos , Luciferasas/análisis , Luciferasas/genética , Mitosis , Biosíntesis de Proteínas , Ratas , Ratas Sprague-Dawley , Transgenes , Proteínas Virales/metabolismo
19.
Cancer Res ; 78(20): 5863-5876, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30154144

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Proteínas de la Membrana/metabolismo , Securina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Anciano , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Reparación del ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Estimación de Kaplan-Meier , Lentivirus/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Infecciones por Papillomavirus/complicaciones , Proto-Oncogenes Mas , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Análisis de Matrices Tisulares , Resultado del Tratamiento
20.
Nucleic Acids Res ; 33(9): e86, 2005 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-15914665

RESUMEN

Synthetic vectors based on reducible polycations consisting of histidine and polylysine residues (HIS RPCs) were evaluated for their ability to deliver nucleic acids. Initial experiments showed that RPC-based vectors with at least 70% histidine content mediated efficient levels of gene transfer without requirement for the endosomolytic agent chloroquine. Significant gene transfer was observed in a range of cell types achieving up to a 5-fold increase in the percentage of transfected cells compared to 25 kDa PEI, a gold standard synthetic vector. In contrast to 25 kDa PEI, HIS RPCs also mediated efficient transfer of other nucleic acids, including mRNA encoding green fluorescent protein in PC-3 cells and siRNA directed against the neurotrophin receptor p75(NTR) in post-mitotic cultures of rat dorsal root ganglion cell neurons. Experiments to elevate intracellular glutathione and linear profiling of cell images captured by multiphoton fluorescent microscopy highlighted that parameters such as the molecular weight and rate of cleavage of HIS RPCs were important factors in determining transfection activity. Altogether, these results demonstrate that HIS RPCs represent a novel and versatile type of vector that can be used for efficient cytoplasmic delivery of a broad range of nucleic acids. This should enable different or a combination of therapeutic strategies to be evaluated using a single type of polycation-based vector.


Asunto(s)
ADN/administración & dosificación , Vectores Genéticos/química , Histidina/química , Polilisina/química , Transfección/métodos , Animales , Células COS , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Cloroquina/farmacología , Vectores Genéticos/toxicidad , Glutatión/metabolismo , Humanos , Polietileneimina/toxicidad , ARN Mensajero/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Ratas , Sales (Química)/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA