40 research outputs found

    The multifunctional FUS, EWS and TAF15 proto-oncoproteins show cell type-specific expression patterns and involvement in cell spreading and stress response

    Get PDF
    Background: FUS, EWS and TAF15 are structurally similar multifunctional proteins that were first discovered upon characterization of fusion oncogenes in human sarcomas and leukemias. The proteins belong to the FET ( previously TET) family of RNA-binding proteins and are implicated in central cellular processes such as regulation of gene expression, maintenance of genomic integrity and mRNA/microRNA processing. In the present study, we investigated the expression and cellular localization of FET proteins in multiple human tissues and cell types. Results: FUS, EWS and TAF15 were expressed in both distinct and overlapping patterns in human tissues. The three proteins showed almost ubiquitous nuclear expression and FUS and TAF15 were in addition present in the cytoplasm of most cell types. Cytoplasmic EWS was more rarely detected and seen mainly in secretory cell types. Furthermore, FET expression was downregulated in differentiating human embryonic stem cells, during induced differentiation of neuroblastoma cells and absent in terminally differentiated melanocytes and cardiac muscle cells. The FET proteins were targeted to stress granules induced by heat shock and oxidative stress and FUS required its RNA-binding domain for this translocation. Furthermore, FUS and TAF15 were detected in spreading initiation centers of adhering cells. Conclusion: Our results point to cell-specific expression patterns and functions of the FET proteins rather than the housekeeping roles inferred from earlier studies. The localization of FET proteins to stress granules suggests activities in translational regulation during stress conditions. Roles in central processes such as stress response, translational control and adhesion may explain the FET proteins frequent involvement in human cancer

    SMAD4 haploinsufficiency in small intestinal neuroendocrine tumors

    Get PDF
    Background: Patients with small intestinal neuroendocrine tumors (SINETs) frequently present with lymph node and liver metastases at the time of diagnosis, but the molecular changes that lead to the progression of these tumors are largely unknown. Sequencing studies have only identified recurrent point mutations at low frequencies with CDKN1B being the most common harboring heterozygous mutations in less than 10% of all tumors. Although SINETs are genetically stable tumors with a low frequency of point mutations and indels, they often harbor recurrent hemizygous copy number alterations (CNAs) yet the functional implications of these CNA are unclear. Methods: Utilizing comparative genomic hybridization (CGH) arrays we analyzed the CNA profile of 131 SINETs from 117 patients. Two tumor suppressor genes and corresponding proteins i.e. SMAD4, and CDKN1B, were further characterized using a tissue microarray (TMA) with 846 SINETs. Immunohistochemistry (IHC) was used to quantify protein expression in TMA samples and this was correlated with chromosome number evaluated with fluorescent in-situ hybridization (FISH). Intestinal tissue from a Smad4+/− mouse model was used to detect entero-endocrine cell hyperplasia with IHC. Results: Analyzing the CGH arrays we found loss of chromosome 18q and SMAD4 in 71% of SINETs and that focal loss of chromosome 12 affecting the CDKN1B was present in 9.4% of SINETs. No homozygous loss of chromosome 18 was detected. Hemizygous loss of SMAD4, but not CDKN1B, significantly correlated with reduced protein levels but hemizygous loss of SMAD4 did not induce entero-endocrine cell hyperplasia in the Smad4+/− mouse model. In addition, patients with low SMAD4 protein expression in primary tumors more often presented with metastatic disease. Conclusions: Hemizygous loss of chromosome 18q and the SMAD4 gene is the most common genetic event in SINETs and our results suggests that this could influence SMAD4 protein expression and spread of metastases. Although SMAD4 haploinsufficiency alone did not induce tumor initiation, loss of chromosome 18 could represent an evolutionary advantage in SINETs explaining the high prevalence of this aberration. Functional consequences of reduced SMAD4 protein levels could hypothetically be a potential mechanism as to why loss of chromosome 18 appears to be clonally selected in SINETs

    177 Lu-octreotate therapy for neuroendocrine tumours is enhanced by Hsp90 inhibition

    Get PDF
    Lu-177-octreotate is an FDA-approved radionuclide therapy for patients with gastroenteropancreatic neuroendocrine tumours (NETs) expressing somatostatin receptors. The Lu-177-octreotate therapy has shown promising results in clinical trials by prolonging progression-free survival, but complete responses are still uncommon. The aim of this study was to improve the Lu-177-octreotate therapy by means of combination therapy. To identify radiosensitising inhibitors, two cell lines, GOT1 and P-STS, derived from small intestinal neuroendocrine tumours (SINETs), were screened with 1224 inhibitors alone or in combination with external radiation. The screening revealed that inhibitors of Hsp90 can potentiate the tumour cell-killing effect of radiation in a synergistic fashion (GOT1; false discovery rate < 3.2 x 10(-11)). The potential for Hsp90 inhibitor ganetespib to enhance the anti-tumour effect of Lu-177-octreotate in an in vivo setting was studied in the somatostatin receptor-expressing GOT1 xenograft model. The combination led to a larger decrease in tumour volume relative to monotherapies and the tumour-reducing effect was shown to be synergistic. Using patient-derived tumour cells from eight metastatic SINETs, we could show that ganetespib enhanced the effect of Lu-177-octreotate therapy for all investigated patient tumours. Levels of Hsp90 protein expression were evaluated in 767 SINETs from 379 patients. We found that Hsp90 expression was upregulated in tumour cells relative to tumour stroma in the vast majority of SINETs. We conclude that Hsp90 inhibitors enhance the tumour-killing effect of Lu-177-octreotate therapy synergistically in SINET tumour models and suggest that this potentially promising combination should be further evaluated

    Döden som katharsis. Nordiska perspektiv pÄ dödens kultur och mentalitetshistoria

    Get PDF
    Döden Ă€r ett evigt tema. Men diskursen runt döden i dess mĂ„ngskiftande former Ă€r underkastad stĂ€ndig förĂ€ndring. Döden kan ses som ett hot och ett gissel men ocksĂ„ som en befrielse, ett straff eller som ett redskap för att befria samhĂ€llet frĂ„n oönskade individer. I samtliga fall kan man tala om döden som en reningsprocess – som katharsis. Det Ă€r denna förĂ€nderliga diskurs som tas upp i denna bok. I centrum stĂ„r frĂ„gan om hur man hanterat dödsproblematiken under olika tider och hur man anvĂ€nt sig av döden som ett redskap i samhĂ€llets tjĂ€nst. Vidare diskuteras hur döden relaterats till individ och samhĂ€lle och vilken funktion samhĂ€llets religiöst eller ideologiskt motiverade vĂ€rdegrund haft i detta sammanhang. Bokens Ă„tta artiklar belyser synen pĂ„ döden i VĂ€sterlandet i ett historiskt perspektiv

    The neuroendocrine phenotype, genomic profile and therapeutic sensitivity of GEPNET cell lines

    Get PDF
    Experimental models of neuroendocrine tumour disease are scarce, and no comprehensive characterisation of existing gastroenteropancreatic neuroendocrine tumour (GEPNET) cell lines has been reported. In this study, we aimed to define the molecular characteristics and therapeutic sensitivity of these cell lines. We therefore performed immunophenotyping, copy number profiling, whole-exome sequencing and a large-scale inhibitor screening of seven GEPNET cell lines. Four cell lines, GOT1, P-STS, BON-1 and QGP-1, displayed a neuroendocrine phenotype while three others, KRJ-I, L-STS and H-STS, did not. Instead, these three cell lines were identified as lymphoblastoid. Characterisation of remaining authentic GEPNET cell lines by copy number rofiling showed that GOT1, among other chromosomal alterations, harboured losses on chromosome 18 encompassing the SMAD4 gene, while P-STS had a loss on 11q. BON-1 had a homozygous loss of CDKN2A and CDKN2B, and QGP-1 harboured amplifications of MDM2 and HMGA2. Whole-exome sequencing revealed both disease-characteristic mutations (e.g. ATRX mutation in QGP-1) and, for patient tumours, rare genetic events (e.g. TP53 mutation in P-STS, BON-1 and QGP-1). A large-scale inhibitor screening showed that cell lines from pancreatic NETs to a greater extent, when compared to small intestinal NETs, were sensitive to inhibitors of MEK. Similarly, neuroendocrine NET cells originating from the small intestine were considerably more sensitive to a group of HDAC inhibitors. Taken together, our results provide a comprehensive characterisation of GEPNET cell lines, demonstrate their relevance as neuroendocrine tumour models and explore their therapeutic sensitivity to a broad range of inhibitors

    Gene therapy of neuroblastoma

    No full text
    Aims: The survival of children with advanced stages of neuroblastoma has not improved significantly during recent years. Therefore additional treatments based on other modalities need to be developed. The objective of this thesis was to investigate the possibility to develop gene therapy of neuroblastoma. It includes studies of the two potential therapeutic genes Apoptosis Signal-regulating Kinase (ASK1) and the suicide gene E. coli Purine Nucleoside Phosphorylase (PNP), neuroblastoma specific therapy, and activation of the immune system.Results: Adenovirus-mediated expression of a constitutively active form of ASK1, promoted apoptosis in two out of four different neuroblastoma cell lines, judged by AnnexinV staining and DNA laddering. ASK1 induced apoptosis by activating the down stream p38MAP kinase signalling pathway which was visualised by western blot analysis. Nevertheless, it was possible to sensitise the unresponsive cell line, NXS2, with the microtubule-interfering agent, paclitaxel. The MASH1-promoter was placed upstream of the luciferase reporter gene or of the E. coli PNP gene in expression vectors. Measuring the luciferase activity, the different sizes of the proximal promoter displayed cell-type specific activity in neuroblastoma cell lines, when compared in non-neuroblastoma cell lines. The shortest MASH1 promoter was combined with the E.coli PNP gene and the toxicity was measured by MTT assay after addition of the prodrug F-araA. The cytotoxicity was 64 to 65 percent in the neuroblastoma cells with low toxicity in the non-neuroblastoma cell lines, relative to the cytotoxicity of the E.coli PNP gene regulated by the strong but non-specific CMV enhancer-promoter. The growth of tumours and the survival of animals challenged with wild type tumour cells were followed in A/J mice, which had been vaccinated with IL-12 and GM-CSF secreting C1300 tumour cells. The survival of animals, subcutaneously injected with the combination of IL-12 and GM-CSF secreting cells was increased compared to vaccination with IL-12 or GM-CSF alone. The immunological responses were analysed by FACS analysis of the splenocytes. The GM-CSF vaccinated animals had a marked increase of activated T helper cells and the GM-CSF and IL-12/GM-CSF groups had an increased fraction of NK1.1 positive cells expressing the NKG2D receptor. The binding of a NKG2D tetramer revealed that the IL-12 and GM-CSF- expressing tumour cells had an upregulated expression of the MHC class I like NKG2D ligands. This is a step forward facilitating detection and eradication of tumours by the NK and NKT cells carrying the NKG2D receptor.Conclusions: Our results show that it is possible to induce apoptosis of neuroblastoma cells in vitro, by ASK1 and E. coli PNP/F-araA, and the MASH1 promoter can be used as a regulatory region for a high and tissue specific expression of the therapeutic gene. Immunotherapy of a syngeneic neuroblastoma mouse model vaccinated with cells secreting IL-12 and GM-CSF increased survival. To further enhance the antitumour effect it would be interesting to combine immunotherapy with apoptosis-inducing gene therapy

    Hypoxia stimulates CXCR4 signalling in ileal carcinoids.

    No full text
    Tumour hypoxia is associated with increased metastatic potential and resistance to radiotherapy and chemotherapy. Ileal carcinoids are usually metastatic at the time of diagnosis and respond poorly to chemotherapy. The aim of this study was to investigate the extent of hypoxia in ileal carcinoids and the response of tumour cells to induced hypoxia. VEGF, CA-IX, HIF-1alpha and HIF-2alpha were studied by immunohistochemistry in biopsies from 24 patients with ileal carcinoids. All hypoxic markers were shown to be highly expressed in localized areas of the tumours irrespective of tumour location or stage. However, HIF-2alpha expression was significantly higher in distant metastases compared to primary tumours in the same patient. Global gene expression profiling of GOT1 carcinoid cells revealed a marked response to hypoxia. Expression of genes related to epithelial-to-mesenchymal transition (EMT) and development was altered including increased expression of the chemokine receptor CXCR4, an important regulator of invasive growth and metastasis formation. High expression of CXCR4 was confirmed by immunohistochemistry in tumour biopsies. Stimulation of GOT1 cells by the CXCR4 ligand CXCL12 (SDF-1) activated the MAPK p42/44 signalling pathway and increased tumour cell migration. We conclude that ileal carcinoids contain hypoxic areas expressing HIF-1alpha, and HIF-2alpha and CXCR4. Signalling through the CXCL12-CXCR4 axis may contribute to the metastatic potential of ileal carcinoids. Targeting of HIFs and/or the CXCR4 signalling pathway may offer new therapeutic strategies for this carcinoid tumour disease

    Expression profiling of small intestinal neuroendocrine tumors identifies subgroups with clinical relevance, prognostic markers and therapeutic targets.

    No full text
    We wanted to define the transcriptome of small intestinal neuroendocrine tumors in order to identify clinically relevant subgroups of tumors, prognostic markers and novel targets for treatment. Genome-wide expression profiling was conducted on tumor biopsies from 33 patients with well-differentiated neuroendocrine tumors of the distal ileum and metastatic disease at the time of diagnosis. Unsupervised hierarchical clustering analysis identified three groups of tumors. The largest group, comprising half of the tumors, was characterized by longer patient survival and higher expression of neuroendocrine markers, including SSTR2. Tumors with higher grade (G2/3) or gain of chromosome 14 were associated with shorter patient survival and increased expression of cell cycle-promoting genes. Pathway analysis predicted the prostaglandin E receptor 2 (PTGER2) as the most significantly activated regulator in tumors of higher grade, whereas Forkhead box M1 (FOXM1) was the most significantly activated regulator in tumors with gain of chromosome 14. Druggable genes identified from expression profiles included clinically proven SSTR2 and also novel targets, for example, receptor tyrosine kinases (RET, FGFR1/3, PDGFRB and FLT1), epigenetic regulators, molecular chaperones and signal transduction molecules. Evaluation of candidate drug targets on neuroendocrine tumors cells (GOT1) showed significant inhibition of tumor cell growth after treatment with tyrosine kinase inhibitors or inhibitors of HDAC, HSP90 and AKT. In conclusion, we have defined the transcriptome of small intestinal neuroendocrine tumors and identified novel subgroups with clinical relevance. We found specific gene expression patterns associated with tumor grade and chromosomal alterations. Our data also suggest novel prognostic biomarkers and therapies for these patients
    corecore