Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Nat Commun ; 15(1): 3736, 2024 May 14.
Article En | MEDLINE | ID: mdl-38744818

The E3 SUMO ligase PIAS2 is expressed at high levels in differentiated papillary thyroid carcinomas but at low levels in anaplastic thyroid carcinomas (ATC), an undifferentiated cancer with high mortality. We show here that depletion of the PIAS2 beta isoform with a transcribed double-stranded RNA-directed RNA interference (PIAS2b-dsRNAi) specifically inhibits growth of ATC cell lines and patient primary cultures in vitro and of orthotopic patient-derived xenografts (oPDX) in vivo. Critically, PIAS2b-dsRNAi does not affect growth of normal or non-anaplastic thyroid tumor cultures (differentiated carcinoma, benign lesions) or cell lines. PIAS2b-dsRNAi also has an anti-cancer effect on other anaplastic human cancers (pancreas, lung, and gastric). Mechanistically, PIAS2b is required for proper mitotic spindle and centrosome assembly, and it is a dosage-sensitive protein in ATC. PIAS2b depletion promotes mitotic catastrophe at prophase. High-throughput proteomics reveals the proteasome (PSMC5) and spindle cytoskeleton (TUBB3) to be direct targets of PIAS2b SUMOylation at mitotic initiation. These results identify PIAS2b-dsRNAi as a promising therapy for ATC and other aggressive anaplastic carcinomas.


Mitosis , Protein Inhibitors of Activated STAT , Humans , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/genetics , Animals , Cell Line, Tumor , Mice , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , RNA Interference , Spindle Apparatus/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Xenograft Model Antitumor Assays , Proteasome Endopeptidase Complex/metabolism , Sumoylation , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Female
3.
Oncogene ; 40(45): 6354-6368, 2021 11.
Article En | MEDLINE | ID: mdl-34588620

It is unclear how loss-of-function germline mutations in the widely-expressed co-chaperone AIP, result in young-onset growth hormone secreting pituitary tumours. The RET receptor, uniquely co-expressed in somatotrophs with PIT1, induces apoptosis when unliganded, while RET supports cell survival when it is bound to its ligand. We demonstrate that at the plasma membrane, AIP is required to form a complex with monomeric-intracellular-RET, caspase-3 and PKCδ resulting in PIT1/CDKN2A-ARF/p53-apoptosis pathway activation. AIP-deficiency blocks RET/caspase-3/PKCδ activation preventing PIT1 accumulation and apoptosis. The presence or lack of the inhibitory effect on RET-induced apoptosis separated pathogenic AIP variants from non-pathogenic ones. We used virogenomics in neonatal rats to demonstrate the effect of mutant AIP protein on the RET apoptotic pathway in vivo. In adult male rats altered AIP induces elevated IGF-1 and gigantism, with pituitary hyperplasia through blocking the RET-apoptotic pathway. In females, pituitary hyperplasia is induced but IGF-1 rise and gigantism are blunted by puberty. Somatotroph adenomas from pituitary-specific Aip-knockout mice overexpress the RET-ligand GDNF, therefore, upregulating the survival pathway. Somatotroph adenomas from patients with or without AIP mutation abundantly express GDNF, but AIP-mutated tissues have less CDKN2A-ARF expression. Our findings explain the tissue-specific mechanism of AIP-induced somatotrophinomas and provide a previously unknown tumorigenic mechanism, opening treatment avenues for AIP-related tumours.


Acromegaly/genetics , Germ-Line Mutation , Gigantism/genetics , Growth Hormone-Secreting Pituitary Adenoma/genetics , Intracellular Signaling Peptides and Proteins/genetics , Acromegaly/metabolism , Animals , Animals, Newborn , Apoptosis , Cell Line , Female , Gene Knockout Techniques , Gigantism/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Male , Mice , Organ Specificity , Proto-Oncogene Proteins c-ret/metabolism , Rats , Signal Transduction
4.
Article En | MEDLINE | ID: mdl-33071961

The RET tyrosine kinase receptor is expressed by the endocrine somatotroph cells of the pituitary where it has important functions regulating survival/apoptosis. However, RET is also expressed by the GPS pituitary stem cells localized in a niche between the adenopituitary and the intermediate lobe. To bind any of its four ligands, RET needs one of four co-receptors called GFRα1-4. It has been previously shown that GFRα1 is expressed by somatotroph cells and acromegaly tumors. GFRα2 was shown to be expressed by pituitary stem cells. GFRα4 was proposed as not expressed in the pituitary. Here we study the RNA and protein expression of the four GFRα co-receptors for RET in rat and human pituitary. The four co-receptors were abundantly expressed at the RNA level both in rat and human pituitary, although GFRα4 was the less abundant. Multiple immunofluorescence for each co-receptor and ß-catenin, a marker of stem cell niche was performed. The four GFRα co-receptors were co-expressed by the GPS cells at the niche colocalizing with ß-catenin. Isolated individual scattered cells positive for one or other receptor could be found through the adenopituitary with low ß-catenin expression. Some of them co-express GFRα1 and PIT1. Immunohistochemistry in normal human pituitary confirmed the data. Our data suggest that the redundancy of GFRα co-expression is a self-supportive mechanism which ensures niche maintenance and proper differentiation.


Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Pituitary Gland/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Stem Cells/metabolism , Animals , Female , Humans , Male , Rats , Stem Cell Niche
5.
EBioMedicine ; 43: 537-552, 2019 May.
Article En | MEDLINE | ID: mdl-30975543

BACKGROUND: Acromegaly is produced by excess growth hormone secreted by a pituitary adenoma of somatotroph cells (ACRO). First-line therapy, surgery and adjuvant therapy with somatostatin analogs, fails in 25% of patients. There is no predictive factor of resistance to therapy. New therapies are investigated using few dispersed tumor cells in acute primary cultures in standard conditions where the cells do not grow, or using rat pituitary cell lines that do not maintain the full somatotroph phenotype. The RET/PIT1/p14ARF/p53 pathway regulates apoptosis in normal pituitary somatotrophs whereas the RET/GDNF pathway regulates survival, controlling PIT1 levels and blocking p14ARF (ARF) and p53 expression. METHODS: We investigated these two RET pathways in a prospective series of 32 ACRO and 63 non-functioning pituitary adenomas (NFPA), studying quantitative RNA and protein gene expression for molecular-clinical correlations and how the RET pathway might be implicated in therapeutic success. Clinical data was collected during post-surgical follow-up. We also established new'humanized' pituitary cultures, allowing 20 repeated passages and maintaining the pituitary secretory phenotype, and tested five multikinase inhibitors (TKI: Vandetanib, Lenvatinib, Sunitinib, Cabozantinib and Sorafenib) potentially able to act on the GDNF-induced RET dimerization/survival pathway. Antibody arrays investigated intracellular molecular pathways. FINDINGS: In ACRO, there was specific enrichment of all genes in both RET pathways, especially GDNF. ARF and GFRA4 gene expression were found to be opposing predictors of response to first-line therapy. ARF cut-off levels, calculated categorizing by GNAS mutation, were predictive of good response (above) or resistance (below) to therapy months later. Sorafenib, through AMPK, blocked the GDNF/AKT survival action without altering the RET apoptotic pathway. INTERPRETATION: Tumor ARF mRNA expression measured at the time of the surgery is a prognosis factor in acromegaly. The RET inhibitor, Sorafenib, is proposed as a potential treatment for resistant ACRO. FUND: This project was supported by national grants from Agencia Estatal de Investigación (AEI) and Instituto Investigación Carlos III, with participation of European FEDER funds, to IB (PI150056) and CVA (BFU2016-76973-R). It was also supported initially by a grant from the Investigator Initiated Research (IIR) Program (WI177773) and by a non-restricted Research Grant from Pfizer Foundation to IB. Some of the pituitary acromegaly samples were collected in the framework of the Spanish National Registry of Acromegaly (REMAH), partially supported by an unrestricted grant from Novartis to the Spanish Endocrine Association (SEEN). CVA is also supported from a grant of Medical Research Council UK MR/M018539/1.


Acromegaly/diagnosis , Acromegaly/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Transcription Factor Pit-1/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism , Acromegaly/genetics , Acromegaly/therapy , Animals , Apoptosis/genetics , Biomarkers , Combined Modality Therapy , Gene Expression Profiling , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Humans , Immunohistochemistry , Models, Biological , Mutation , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Prognosis , Proto-Oncogene Proteins c-ret/genetics , Rats , Signal Transduction , Transcription Factor Pit-1/genetics , Treatment Outcome , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics
6.
J Clin Endocrinol Metab ; 98(6): 2431-41, 2013 Jun.
Article En | MEDLINE | ID: mdl-23539720

CONTEXT: Mechanisms of thyroid physiology and cancer are principally studied in follicular cell lines. However, human thyroid cancer lines were found to be heavily contaminated by other sources, and only one supposedly normal-thyroid cell line, immortalized with SV40 antigen, is available. In primary culture, human follicular cultures lose their phenotype after passage. We hypothesized that the loss of the thyroid phenotype could be related to culture conditions in which human cells are grown in medium optimized for rodent culture, including hormones with marked differences in its affinity for the relevant rodent/human receptor. OBJECTIVE: The objective of the study was to define conditions that allow the proliferation of primary human follicular thyrocytes for many passages without losing phenotype. METHODS: Concentrations of hormones, transferrin, iodine, oligoelements, antioxidants, metabolites, and ethanol were adjusted within normal homeostatic human serum ranges. Single cultures were identified by short tandem repeats. Human-rodent interspecies contamination was assessed. RESULTS: We defined an humanized 7 homeostatic additives medium enabling growth of human thyroid cultures for more than 20 passages maintaining thyrocyte phenotype. Thyrocytes proliferated and were grouped as follicle-like structures; expressed Na+/I- symporter, pendrin, cytokeratins, thyroglobulin, and thyroperoxidase showed iodine-uptake and secreted thyroglobulin and free T3. Using these conditions, we generated a bank of thyroid tumors in culture from normal thyroids, Grave's hyperplasias, benign neoplasms (goiter, adenomas), and carcinomas. CONCLUSIONS: Using appropriate culture conditions is essential for phenotype maintenance in human thyrocytes. The bank of thyroid tumors in culture generated under humanized humanized 7 homeostatic additives culture conditions will provide a much-needed tool to compare similarly growing cells from normal vs pathological origins and thus to elucidate the molecular basis of thyroid disease.


Cell Culture Techniques , Culture Media , Thyroid Gland/cytology , Thyroid Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Phenotype , Rats , Thyroglobulin/metabolism , Triiodothyronine/metabolism
...