Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Small ; 19(1): e2203331, 2023 01.
Article in English | MEDLINE | ID: mdl-36403214

ABSTRACT

Nanocrystalline ZnO sponges doped with 5 mol% EuO1.5 are obtained by heating metal-salt complex based precursor pastes at 200-900 °C for 3 min. X-ray diffraction, transmission electron microscopy, and extended X-ray absorption fine structure (EXAFS) show that phase separation into ZnO:Eu and c-Eu2 O3 takes place upon heating at 700 °C or higher. The unit cell of the clean oxide made at 600 °C shows only ≈0.4% volume increase versus undoped ZnO, and EXAFS shows a ZnO local structure that is little affected by the Eu-doping and an average Eu3+ ion coordination number of ≈5.2. Comparisons of 23 density functional theory-generated structures having differently sized Eu-oxide clusters embedded in ZnO identify three structures with four or eight Eu atoms as the most energetically favorable. These clusters exhibit the smallest volume increase compared to undoped ZnO and Eu coordination numbers of 5.2-5.5, all in excellent agreement with experimental data. ZnO defect states are crucial for efficient Eu3+ excitation, while c-Eu2 O3 phase separation results in loss of the characteristic Eu3+ photoluminescence. The formation of molecule-like Eu-oxide clusters, entrapped in ZnO, proposed here, may help in understanding the nature of the unexpected high doping levels of lanthanide ions in ZnO that occur virtually without significant change in ZnO unit cell dimensions.


Subject(s)
Lanthanoid Series Elements , Zinc Oxide , Zinc Oxide/chemistry , Europium/chemistry , X-Ray Diffraction
2.
Sci Rep ; 11(1): 22733, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34815475

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs) are slow-growing tumors that seem genetically quite stable without highly recurrent mutations, but are epigenetically dysregulated. In contrast to the undetectable expression of the enhancer of zeste homolog 2 (EZH2) histone methyltransferase in the enterochromaffin cells of the small intestine, we found high and differential expression of EZH2 in primary SI-NETs and corresponding metastases. Silencing EZH2 in the SI-NET cell line CNDT2.5 reduced cell proliferation and induced apoptosis. Furthermore, EZH2 knockout inhibited tumor progression in a CNDT2.5 SI-NET xenograft mouse model, and treatment of SI-NET cell lines CNDT2.5 and GOT1 with the EZH2-specific inhibitor CPI-1205 decreased cell viability and promoted apoptosis. Moreover, CPI-1205 treatment reduced migration capacity of CNDT2.5 cells. The EZH2 inhibitor GSK126 also repressed proliferation of CNDT2.5 cells. Recently, metformin has received wide attention as a therapeutic option in diverse cancers. In CNDT2.5 and GOT1 cells, metformin suppressed EZH2 expression, and inhibited cell proliferation. Exposure of GOT1 three-dimensional cell spheroids to CPI-1205 or metformin arrested cell proliferation and decreased spheroid size. These novel findings support a possible role of EZH2 as a candidate oncogene in SI-NETs, and suggest that CPI-1205 and metformin should be further evaluated as therapeutic options for patients with SI-NETs.


Subject(s)
Biomarkers, Tumor/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Indoles/pharmacology , Intestinal Neoplasms/drug therapy , Intestine, Small/drug effects , Neuroendocrine Tumors/drug therapy , Piperidines/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Female , Humans , Hypoglycemic Agents/pharmacology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Metformin/pharmacology , Mice , Mice, Nude , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Inorg Chem ; 59(11): 7584-7602, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32374596

ABSTRACT

A low-cost template-free solution chemical route to highly porous nanocrystalline sponges of ZnO-EuO1.5 with 0-5 mol % Eu is presented. The process uses Zn- and Eu-acetate-nitrate and triethanolamine as precursors in methanol. After evaporation of the solvent and heating at 200 °C for 3 min, crystalline ZnO:Eu sponges with minor amounts of organic residues were obtained. Heating to 400 °C replaced the organics with carbonate, which in its turn was decomposed at temperatures below 600 °C, forming ZnO:Eu sponges. Samples heated to 200-1000 °C for 3 min were studied with XRD, SEM, TEM, TG, XPS, and IR spectroscopy. The ZnO:Eu crystallite sizes could be tuned from below 10 nm for sponges prepared at 200-500 °C, to over 100 nm range at 900 °C, without sintering of the overall microstructure. XRD showed the presence of hexagonal ZnO:Eu (or at 700-1000 °C, ZnO:Eu and cubic Eu2O3) as the only phases present. The ZnO:Eu had slightly larger unit cell dimensions than the literature value of ZnO for samples obtained at 200-600 °C, while the unit cells of samples obtained at higher temperatures were quite close to the value of undoped ZnO. XPS showed that Eu was mainly in its 3+ state and well-distributed within the sponges but segregated at the ZnO sponge surface upon heating at 700-1000 °C, in accordance with XRD studies showing Eu2O3 formation.

4.
Endocr Connect ; 8(8): 1126-1135, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31349215

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs) are small, slow growing neoplasms with loss of one copy of chromosome 18 as a common event. Frequently mutated genes on chromosome 18 or elsewhere have not been found so far. The aim of this study was to investigate a possible tumor suppressor role of the transmembrane receptor type tyrosine phosphatase PTPµ (PTPRM at 18p11) in SI-NETs. Immunohistochemistry, quantitative RT-PCR, colony formation assay and quantitative CpG methylation analysis by pyrosequencing were performed. Undetectable/very low levels of PTPRM or aberrant pattern of immunostaining, with both negative and positive areas, were detected in the majority of tumors (33/40), and a significantly reduced mRNA expression in metastases compared to primary tumors was observed. Both the DNA methylation inhibitor 5-aza-2'-deoxycytidine and the S-adenosylhomocysteine hydrolase inhibitor 3-deazaneplanocin A (DZNep) induced PTPRM expression in CNDT2.5 and KRJ-I SI-NET cells. CpG methylation of upstream regulatory regions, the promoter region and the exon 1/intron 1 boundary was detected by pyrosequencing analysis of the two cell lines and not in the analyzed SI-NETs. Overexpression of PTPRM in the SI-NET cell lines reduced cell growth and cell proliferation and induced apoptosis. The tyrosine phosphatase activity of PTPRM was not involved in cell growth inhibition. The results support a role for PTPRM as a dysregulated candidate tumor suppressor gene in SI-NETs and further analyses of the involved mechanisms are warranted.

5.
ChemSusChem ; 12(2): 404-408, 2019 Jan 24.
Article in English | MEDLINE | ID: mdl-30485687

ABSTRACT

Herein, a catalytic reductive fractionation of lignocellulose is presented using a heterogeneous cobalt catalyst and formic acid or formate as a hydrogen donor. The catalytic reductive fractionation of untreated birch wood yields monophenolic compounds in up to 34 wt % yield of total lignin, which corresponds to 76 % of the theoretical maximum yield. Model compound studies revealed that the main role of the cobalt catalyst is to stabilize the reactive intermediates formed during the organosolv pulping by transfer hydrogenation and hydrogenolysis reactions. Additionally, the cobalt catalyst is responsible for depolymerization reactions of lignin fragments through transfer hydrogenolysis reactions, which target the ß-O-4' bond. The catalyst could be recycled three times with only negligible decrease in efficiency, showing the robustness of the system.

6.
BMC Cancer ; 18(1): 764, 2018 Jul 25.
Article in English | MEDLINE | ID: mdl-30045709

ABSTRACT

BACKGROUND: Small intestinal neuroendocrine tumors (SI-NETs) originate from enterochromaffin cells scattered in the intestinal mucosa of the ileum and jejunum. Loss of one copy of chromosome 18 is the most frequent observed aberration in primary tumors and metastases. The aim of this study was to investigate possible involvement of 5-hydroxymethylcytosine (5hmC), TET1 and TET2 in SI-NETs. METHODS: The analysis was conducted using 40 primary tumors and corresponding 47 metastases. The level of 5hmC, TET1 and TET2 was analyzed by DNA immune-dot blot assay and immunohistochemistry. Other methods included a colony forming assay, western blotting analysis, and quantitative bisulfite pyrosequencing analysis. The effect of the exportin-1 nuclear transport machinery inhibitors on cell proliferation and apoptosis was also explored using two SI-NET cell lines. RESULTS: Variable levels of 5hmC and a mosaic staining appearance with a mixture of positive and negative cell nuclei, regardless of cell number and staining strength, was observed overall both in primary tumors and metastases. Similarly aberrant staining pattern was observed for TET1 and TET2. In a number of tumors (15/32) mosaic pattern together with areas of negative staining was also observed for TET1. Abolished expression of TET1 in the tumors did not seem to involve hypermethylation of the TET1 promoter region. Overexpression of TET1 in a colony forming assay supported a function as cell growth regulator. In contrast to 5hmC and TET1, TET2 was also observed in the cytoplasm of all the analyzed SI-NETs regardless of nuclear localization. Treatment of CNDT2.5 and KRJ-I cells with the exportin-1 (XPO1/CRM1) inhibitor, leptomycin B, induced reduction in the cytoplasm and nuclear retention of TET2. Aberrant partitioning of TET2 from the nucleus to the cytoplasm seemed therefore to involve the exportin-1 nuclear transport machinery. Reduced cell proliferation and induction of apoptosis were observed after treatment of CNDT2.5 and KRJ-I cells with leptomycin B or KPT-330 (selinexor). CONCLUSIONS: SI-NETs are epigenetically dysregulated at the level of 5-hydroxymethylcytosine/ TET1/TET2. We suggest that KPT-330/selinexor or future developments should be considered and evaluated for single treatment of patients with SI-NET disease and also in combinations with somatostatin analogues, peptide receptor radiotherapy, or everolimus.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Intestinal Neoplasms/metabolism , Mixed Function Oxygenases/metabolism , Neuroendocrine Tumors/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analysis , 5-Methylcytosine/metabolism , Adult , Aged , Cell Nucleus/chemistry , Cell Nucleus/metabolism , DNA-Binding Proteins/analysis , Dioxygenases , Humans , Intestinal Neoplasms/chemistry , Intestine, Small/chemistry , Intestine, Small/metabolism , Middle Aged , Mixed Function Oxygenases/analysis , Neuroendocrine Tumors/chemistry , Proto-Oncogene Proteins/analysis
7.
Inorg Chem ; 56(24): 15150-15158, 2017 Dec 18.
Article in English | MEDLINE | ID: mdl-29172508

ABSTRACT

An efficient, template-free solution-chemical route to nanostructured ZnO sponges is presented: A mixture of Zn(NO3)2·6H2O, Zn(OAc)2·2H2O, and triethanolamine in methanol was evaporated to a highly viscous liquid and rapidly heated to >200 °C for 1-3 min to achieve highly porous, nanocrystalline sponges of ZnO. The viscous precursor concentrate obtained on evaporation in air was characterized by TG, DSC, and IR spectroscopy, and the product ZnO sponges by XRD, SEM, TEM, and IR spectroscopy. The fast reaction forming ZnO started at 140 °C and finished within a few seconds. Scherrer analysis of the XRD peak broadening showed average crystallite sizes of 8 to 11 nm for ZnO prepared by annealing at 200-450 °C (3 min), while grain growth to 134 nm was observed from 500 to 900 °C (3 min). The ZnO powders obtained at 200-900 °C had cell dimensions of a = 3.25 Å and b = 5.21 Å, matching the ZnO literature data well. SEM and TEM analyses showed highly porous, bread-like 3D nanostructures built by ca. 30-70 nm thick walls of ZnO crystallites of the approximate average sizes given by the XRD Scherrer analysis. It seems that the crystal growth above 450 °C takes place within the ZnO 3D structure obtained at lower temperatures without much sintering of the larger porous structure.

8.
Endocr Relat Cancer ; 24(7): 329-338, 2017 07.
Article in English | MEDLINE | ID: mdl-28642344

ABSTRACT

Primary hyperparathyroidism (pHPT) is rarely caused by parathyroid carcinoma (PC, <1-5% of pHPT cases). The TET proteins oxidize the epigenetic mark 5-methylcytosine to 5-hydroxymethylcytosine (5hmC) and inactivation by mutation or epigenetic deregulation of TET1 and TET2 play important roles in various cancers. Recently, we found that 5hmC was severely reduced in all of the analyzed PCs and with deranged expression of TET1 for the majority of PCs. Here, we have examined the expression of the TET2 protein in 15 5hmC-negative PCs from patients who had local invasion or metastases. Cell growth and cell migratory roles for TET2 as well as epigenetic deregulated expression were addressed. Immunohistochemistry revealed very low/undetectable expression of TET2 in all PCs and verified for two PCs that were available for western blotting analysis. Knockdown of TET2 in the parathyroid cell line sHPT-1 resulted in increased cell growth and increased cell migration. DNA sequencing of TET2 in PCs revealed two common variants and no obvious inactivating mutations. Quantitative bisulfite pyrosequencing analysis of the TET2 promoter CpG island revealed higher CpG methylation level in the PCs compared to that in normal tissues and treatment of a PC primary cell culture with the DNA methylation inhibitor 5-aza-2'-deoxycytidine caused increased expression of the methylated TET2 gene. Hence, the data suggest that deregulated expression of TET2 by DNA hypermethylation may contribute to the aberrantly low level of 5hmC in PCs and further that TET2 plays a cell growth and cell migratory regulatory role and may constitute a parathyroid tumor suppressor gene.


Subject(s)
DNA-Binding Proteins/genetics , Parathyroid Neoplasms/genetics , Proto-Oncogene Proteins/genetics , 5-Methylcytosine/metabolism , Cell Growth Processes/genetics , Cell Movement/genetics , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases , Genes, Tumor Suppressor , Humans , Parathyroid Neoplasms/metabolism , Parathyroid Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Transfection
9.
Neuroendocrinology ; 105(2): 170-181, 2017.
Article in English | MEDLINE | ID: mdl-27829249

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs) are amine- and peptide-producing neoplasms. Most patients display metastases at the time of diagnosis; they have an unpredictable individual disease course and the tumors are often therapy resistant. Chromogranin A and 5-hydroxyindoleacetic acid are the biomarkers clinically used most often today, but there is a great need for novel diagnostic and prognostic biomarkers and new therapeutic targets. Sixty-nine biomarkers were screened in serum from 23 SI-NET patients and 23 healthy controls using the multiplex proximity ligation assay (PLA). A refined method, the proximity extension assay (PEA), was used to analyze 76 additional biomarkers. Statistical testing and multivariate classification were performed. Immunohistochemistry and ELISA were performed in an extended cohort. Using PLA, 19 biomarkers showed a significant difference in serum concentrations between patients and controls, and PEA revealed a difference in the concentrations of 17 proteins. Multivariate classification analysis revealed decoy receptor 3 (DcR3), trefoil factor 3 (TFF3), and midkine to be good biomarkers for the disease, which was confirmed by ELISA analysis. All 3 biomarkers were expressed in tumor tissue. DcR3 concentrations were elevated in patients with stage IV disease. High concentrations of DcR3 and TFF3 were correlated to poor survival. DcR3, TFF3, and midkine exhibited elevated serum concentrations in SI-NET patients compared to healthy controls, and DcR3 and TFF3 were associated with poor survival. DcR3 seems to be a marker for liver metastases, while TFF3 and midkine may be new diagnostic biomarkers for SI-NETs.


Subject(s)
Cytokines/blood , Intestinal Neoplasms/blood , Neuroendocrine Tumors/blood , Receptors, Tumor Necrosis Factor, Member 6b/blood , Trefoil Factor-3/blood , Biomarkers, Tumor/blood , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Immunohistochemistry , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Liver Neoplasms/blood , Liver Neoplasms/secondary , Male , Middle Aged , Midkine , Multivariate Analysis , Neoplasm Staging , Neuroendocrine Tumors/pathology , Prognosis , Survival Analysis
10.
BMC Endocr Disord ; 16: 19, 2016 Apr 23.
Article in English | MEDLINE | ID: mdl-27107594

ABSTRACT

BACKGROUND: Small intestinal neuroendocrine tumors (SI-NETs) originate from the enterochromaffin cells in the ileum and jejunum. The knowledge about genetic and epigenetic abnormalities is limited. Low mRNA expression levels of actin gamma smooth muscle 2 (ACTG2) have been demonstrated in metastases relative to primary SI-NETs. ACTG2 and microRNA-145 (miR-145) are aberrantly expressed in other cancers and ACTG2 can be induced by miR-145. The aim of this study was to investigate the role of ACTG2 in small intestinal neuroendocrine tumorigenesis. METHODS: Protein expression was analyzed in SI-NETs (n = 24) and in enterochromaffin cells by immunohistochemistry. The cell line CNDT2.5 was treated with the histone methyltransferase inhibitor 3-deazaneplanocin A (DZNep), the selective EZH2 inhibitor EPZ-6438, or 5-aza-2'-deoxycytidine, a DNA hypomethylating agent. Cells were transfected with ACTG2 expression plasmid or miR-145. Western blotting analysis, quantitative RT-PCR, colony formation- and viability assays were performed. miR-145 expression levels were measured in tumors. RESULTS: Eight primary tumors and two lymph node metastases displayed variable levels of positive staining. Fourteen SI-NETs and normal enterochromaffin cells stained negatively. Overexpression of ACTG2 significantly inhibited CNDT2.5 cell growth. Treatment with DZNep or transfection with miR-145 induced ACTG2 expression (>10-fold), but no effects were detected after treatment with EPZ-6438 or 5-aza-2'-deoxycytidine. DZNep also induced miR-145 expression. SI-NETs expressed relatively low levels of miR-145, with reduced expression in metastases compared to primary tumors. CONCLUSIONS: ACTG2 is expressed in a fraction of SI-NETs, can inhibit cell growth in vitro, and is positively regulated by miR-145. Theoretical therapeutic strategies based on these results are discussed.


Subject(s)
Actins/physiology , Carcinogenesis/genetics , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Actins/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Enterochromaffin Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intestinal Neoplasms/pathology , Intestine, Small/pathology , MicroRNAs/physiology , Middle Aged , Neuroendocrine Tumors/pathology , Young Adult
11.
Clin Epigenetics ; 8: 31, 2016.
Article in English | MEDLINE | ID: mdl-26973719

ABSTRACT

BACKGROUND: Primary hyperparathyroidism is characterized by enlarged parathyroid glands due to an adenoma (80-85 %) or multiglandular disease (~15 %) causing hypersecretion of parathyroid hormone (PTH) and generally hypercalcemia. Parathyroid cancer is rare (<1-5 %). The epigenetic mark 5-hydroxymethylcytosine (5hmC) is reduced in various cancers, and this may involve reduced expression of the ten-eleven translocation 1 (TET1) enzyme. Here, we have performed novel experiments to determine the 5hmC level and TET1 protein expression in 43 parathyroid adenomas (PAs) and 17 parathyroid carcinomas (PCs) from patients who had local invasion or metastases and to address a potential growth regulatory role of TET1. RESULTS: The global 5hmC level was determined by a semi-quantitative DNA immune-dot blot assay in a smaller number of tumors. The global 5hmC level was reduced in nine PCs and 15 PAs compared to four normal tissue samples (p < 0.05), and it was most severely reduced in the PCs. By immunohistochemistry, all 17 PCs stained negatively for 5hmC and TET1 showed negative or variably heterogeneous staining for the majority. All 43 PAs displayed positive 5hmC staining, and a similar aberrant staining pattern of 5hmC and TET1 was seen in about half of the PAs. Western blotting analysis of two PCs and nine PAs showed variable TET1 protein expression levels. A significantly higher tumor weight was associated to PAs displaying a more severe aberrant staining pattern of 5hmC and TET1. Overexpression of TET1 in a colony forming assay inhibited parathyroid tumor cell growth. CONCLUSIONS: 5hmC can discriminate between PAs and PCs. Whether 5hmC represents a novel marker for malignancy warrants further analysis in additional parathyroid tumor cohorts. The results support a growth regulatory role of TET1 in parathyroid tissue.


Subject(s)
Adenoma/chemistry , Cytosine/analogs & derivatives , Parathyroid Neoplasms/chemistry , 5-Methylcytosine/analogs & derivatives , Adenoma/enzymology , Adolescent , Adult , Aged , Blotting, Western , Case-Control Studies , Cytosine/analysis , DNA-Binding Proteins/metabolism , Humans , Middle Aged , Mixed Function Oxygenases , Parathyroid Glands/chemistry , Parathyroid Glands/enzymology , Parathyroid Neoplasms/enzymology , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , Young Adult
12.
Endocr Relat Cancer ; 22(5): 735-44, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285814

ABSTRACT

Aldosterone-producing adenomas (APAs) are found in 1.5-3.0% of hypertensive patients in primary care and can be cured by surgery. Elucidation of genetic events may improve our understanding of these tumors and ultimately improve patient care. Approximately 40% of APAs harbor a missense mutation in the KCNJ5 gene. More recently, somatic mutations in CACNA1D, ATP1A1 and ATP2B3, also important for membrane potential/intracellular Ca(2) (+) regulation, were observed in APAs. In this study, we analyzed 165 APAs for mutations in selected regions of these genes. We then correlated mutational findings with clinical and molecular phenotype using transcriptome analysis, immunohistochemistry and semiquantitative PCR. Somatic mutations in CACNA1D in 3.0% (one novel mutation), ATP1A1 in 6.1% (six novel mutations) and ATP2B3 in 3.0% (two novel mutations) were detected. All observed mutations were located in previously described hotspot regions. Patients with tumors harboring mutations in CACNA1D, ATP1A1 and ATP2B3 were operated at an older age, were more often male and had tumors that were smaller than those in patients with KCNJ5 mutated tumors. Microarray transcriptome analysis segregated KCNJ5 mutated tumors from ATP1A1/ATP2B3 mutated tumors and those without mutation. We observed significant transcription upregulation of CYP11B2, as well as the previously described glomerulosa-specific gene NPNT, in ATP1A1/ATP2B3 mutated tumors compared to KCNJ5 mutated tumors. In summary, we describe novel somatic mutations in proteins regulating the membrane potential/intracellular Ca(2) (+) levels, and also a distinct mRNA and clinical signature, dependent on genetic alteration.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Aldosterone/metabolism , Biomarkers, Tumor/genetics , Hyperaldosteronism/genetics , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/pathology , Adrenocortical Adenoma/metabolism , Adrenocortical Adenoma/pathology , Adult , Aged , Aged, 80 and over , Calcium Channels, L-Type/genetics , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Hyperaldosteronism/metabolism , Hyperaldosteronism/pathology , Immunoenzyme Techniques , Male , Middle Aged , Mutation , Neoplasm Staging , Phenotype , Plasma Membrane Calcium-Transporting ATPases/genetics , Polymerase Chain Reaction , Prognosis , Sodium-Potassium-Exchanging ATPase/genetics
13.
Proc Natl Acad Sci U S A ; 112(13): 4062-7, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25787250

ABSTRACT

Insulinomas are pancreatic islet tumors that inappropriately secrete insulin, producing hypoglycemia. Exome and targeted sequencing revealed that 14 of 43 insulinomas harbored the identical somatic mutation in the DNA-binding zinc finger of the transcription factor Yin Yang 1 (YY1). Chromatin immunoprecipitation sequencing (ChIP-Seq) showed that this T372R substitution changes the DNA motif bound by YY1. Global analysis of gene expression demonstrated distinct clustering of tumors with and without YY1(T372R) mutations. Genes showing large increases in expression in YY1(T372R) tumors included ADCY1 (an adenylyl cyclase) and CACNA2D2 (a Ca(2+) channel); both are expressed at very low levels in normal ß-cells and show mutation-specific YY1 binding sites. Both gene products are involved in key pathways regulating insulin secretion. Expression of these genes in rat INS-1 cells demonstrated markedly increased insulin secretion. These findings indicate that YY1(T372R) mutations are neomorphic, resulting in constitutive activation of cAMP and Ca(2+) signaling pathways involved in insulin secretion.


Subject(s)
Gene Expression Regulation , Insulinoma/genetics , Mutation, Missense , Pancreatic Neoplasms/genetics , YY1 Transcription Factor/genetics , Adenylyl Cyclases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Binding Sites , Blood Glucose/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cohort Studies , Cyclic AMP/metabolism , Female , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulinoma/metabolism , Male , Middle Aged , Molecular Sequence Data , Pancreatic Neoplasms/metabolism , Protein Binding , YY1 Transcription Factor/metabolism
14.
Ann Surg Oncol ; 22 Suppl 3: S1428-35, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25586243

ABSTRACT

BACKGROUND: Until recently, the genetic landscape of small intestinal neuroendocrine tumors (SI-NETs) was limited to recurrent copy number alterations, most commonly a loss on chromosome 18. Intertumor heterogeneity with nonconcordant genotype in paired primary and metastatic lesions also is described, further contributing to the difficulty of unraveling the genetic enigma of SI-NETs. A recent study analyzing 55 SI-NET exomes nominated CDKN1B (p27) as a haploinsufficient tumor suppressor gene. METHODS: This study aimed to determine the frequency of CDKN1B inactivation and to investigate genotype-phenotype correlations. It investigated 362 tumors from 200 patients. All samples were resequenced for mutations in CDKN1B using automated Sanger sequencing. The expression of p27 was investigated in 12 CDKN1B mutant and nine wild type tumors. RESULTS: Some 8.5 % (17/200) of patients had tumors with pathogenic mutations in CDKN1B including 13 insertion deletions, four nonsense variants, and one stop-loss variant. All variants with available nontumoral DNA were classified as somatic. Inter- and intratumor heterogeneity at the CDKN1B locus was detected respectively in six of ten and two of ten patients. Patients with CDKN1B mutated tumors had both heterogeneous disease presentation and diverse prognosis. Expression of the p27 protein did not correlate with CDKN1B mutation status, and no differences in the clinical characteristics between CDKN1B mutated and CDKN1B wild type tumor carriers were found. CONCLUSION: This study corroborates the finding of CDKN1B as a potential haplo-insufficient tumor suppressor gene characterized by inter- and intratumor heterogeneity in SI-NETs.


Subject(s)
Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Intestinal Neoplasms/genetics , Intestine, Small/metabolism , Mutation/genetics , Neuroendocrine Tumors/genetics , Adult , Aged , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Follow-Up Studies , Genetic Heterogeneity , Humans , Immunoenzyme Techniques , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Neuroendocrine Tumors/pathology , Prognosis , Retrospective Studies , Survival Rate
15.
Surgery ; 156(6): 1512-21; discussion 1521-2, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456945

ABSTRACT

BACKGROUND: One-fifth of all patients with small-intestinal neuroendocrine tumors (SI-NETs) present with or develop peritoneal carcinomatosis (PC). Our aim was to determine the prognosis and genetic profiles of tumors in patients with PC compared with tumors in patients without PC. METHODS: We included SI-NET patients (cases with PC, n = 73, and controls without PC, n = 468) who underwent operation between 1985 and 2012. The Lyon prognostic index was used to correlate the amount of PC to survival. DNA samples from patients with (n = 8) and without (n = 7) PC were analyzed with a single-nucleotide polymorphism array (HumanOmni2.5 BeadChip, Illumina) to investigate genetic disparities between groups. RESULTS: Patients with PC had poorer survival (median 5.1 years) than controls (11.1 years). An advanced postoperative Lyon prognostic index was a negative prognostic marker for survival by multivariable analysis (P = .042). Patients with and without PC clustered differently based on loss of heterozygosity and copy number variation data from single-nucleotide polymorphism array of the primary tumors (P = .042). CONCLUSION: SI-NET patients with PC have poor survival, which diminishes with increasing PC load after surgery. Clustering based on copy number variation and loss of heterozygosity data suggests different genotypes in primary tumors comparing patients with and without PC.


Subject(s)
Carcinoma/secondary , Genetic Variation , Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Peritoneal Neoplasms/secondary , Adult , Aged , Carcinoma/genetics , Carcinoma/mortality , Cohort Studies , DNA Copy Number Variations , Disease-Free Survival , Female , Humans , Intestinal Neoplasms/mortality , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Male , Middle Aged , Multivariate Analysis , Neuroendocrine Tumors/mortality , Neuroendocrine Tumors/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/mortality , Prognosis , Reference Values , Retrospective Studies , Risk Assessment , Survival Analysis
17.
Endocr Relat Cancer ; 21(2): 275-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24351681

ABSTRACT

Small intestinal neuroendocrine tumors (SI-NETs), formerly known as midgut carcinoids, are rare and slow-growing neoplasms. Frequent loss of one copy of chromosome 18 in primary tumors and metastases has been observed. The aim of the study was to investigate a possible role of TCEB3C (Elongin A3), currently the only imprinted gene on chromosome 18, as a tumor suppressor gene in SI-NETs, and whether its expression is epigenetically regulated. Primary tumors, metastases, the human SI-NET cell line CNDT2.5, and two other cell lines were included. Immunohistochemistry, gene copy number determination by PCR, colony formation assay, western blotting, real-time quantitative RT-PCR, RNA interference, and quantitative CpG methylation analysis by pyrosequencing were performed. A large majority of tumors (33/43) showed very low to undetectable Elongin A3 expression and as expected 89% (40/45) displayed one gene copy of TCEB3C. The DNA hypomethylating agent 5-aza-2'-deoxycytidine induced TCEB3C expression in CNDT2.5 cells, in primary SI-NET cells prepared directly after surgery, but not in two other cell lines. Also siRNA to DNMT1 and treatment with the general histone methyltransferase inhibitor 3-deazaneplanocin A induced TCEB3C expression in a cell type-specific way. CpG methylation at the TCEB3C promoter was observed in all analyzed tissues and thus not related to expression. Overexpression of TCEB3C resulted in a 50% decrease in clonogenic survival of CNDT2.5 cells, but not of control cells. The results support a putative role of TCEB3C as a tumor suppressor gene in SI-NETs. Epigenetic repression of TCEB3C seems to be tumor cell type-specific and involves both DNA and histone methylation.


Subject(s)
Intestinal Neoplasms/genetics , Neuroendocrine Tumors/genetics , Transcription Factors/genetics , Cell Line, Tumor , CpG Islands , Elongin , Genes, Tumor Suppressor , Histones/metabolism , Humans , Intestinal Neoplasms/metabolism , Intestine, Small , Methylation , Neuroendocrine Tumors/metabolism , Promoter Regions, Genetic
18.
Endocr Relat Cancer ; 21(2): 231-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24292603

ABSTRACT

Primary hyperparathyroidism (pHPT) resulting from parathyroid tumors is a common endocrine disorder with incompletely understood etiology. In renal failure, secondary hyperparathyroidism (sHPT) occurs with multiple tumor development as a result of calcium and vitamin D regulatory disturbance. The aim of this study was to investigate a potential role of the histone 3 lysine 27 methyltransferase EZH2 in parathyroid tumorigenesis. Parathyroid tumors from patients with pHPT included adenomas and carcinomas. Hyperplastic parathyroid glands from patients with HPT secondary to uremia and normal parathyroid tissue specimens were included in this study. Quantitative RT-PCR, western blotting, bisulfite pyrosequencing, colony formation assay, and RNA interference were used. EZH2 was overexpressed in a subset of the benign and in all malignant parathyroid tumors as determined by quantitative RT-PCR and western blotting analyses. Overexpression was explained by EZH2 gene amplification in a large fraction of tumors. EZH2 depletion by RNA interference inhibited sHPT-1 parathyroid cell line proliferation as determined by tritium-thymidine incorporation and colony formation assays. EZH2 depletion also interfered with the Wnt/ß-catenin signaling pathway by increased expression of growth-suppressive AXIN2, a negative regulator of ß-catenin stability. Indeed, EZH2 contributed to the total level of aberrantly accumulated transcriptionally active (nonphosphoylated) ß-catenin in the parathyroid tumor cells. To our knowledge EZH2 gene amplification presents the first genetic aberration common to parathyroid adenomas, secondary hyperplastic parathyroid glands, and parathyroid carcinomas. This supports the possibility of a common pathway in parathyroid tumor development.


Subject(s)
Adenoma/genetics , Carcinoma/genetics , Hyperparathyroidism, Secondary/genetics , Parathyroid Glands/metabolism , Parathyroid Neoplasms/genetics , Polycomb Repressive Complex 2/genetics , Adenoma/metabolism , Carcinoma/metabolism , Enhancer of Zeste Homolog 2 Protein , Humans , Oncogenes , Parathyroid Neoplasms/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Messenger/metabolism , Wnt Signaling Pathway
19.
Nat Genet ; 45(9): 1050-4, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23913001

ABSTRACT

Adrenal aldosterone-producing adenomas (APAs) constitutively produce the salt-retaining hormone aldosterone and are a common cause of severe hypertension. Recurrent mutations in the potassium channel gene KCNJ5 that result in cell depolarization and Ca(2+) influx cause ∼40% of these tumors. We identified 5 somatic mutations (4 altering Gly403 and 1 altering Ile770) in CACNA1D, encoding a voltage-gated calcium channel, among 43 APAs without mutated KCNJ5. The altered residues lie in the S6 segments that line the channel pore. Both alterations result in channel activation at less depolarized potentials; Gly403 alterations also impair channel inactivation. These effects are inferred to cause increased Ca(2+) influx, which is a sufficient stimulus for aldosterone production and cell proliferation in adrenal glomerulosa. We also identified de novo germline mutations at identical positions in two children with a previously undescribed syndrome featuring primary aldosteronism and neuromuscular abnormalities. These findings implicate gain-of-function Ca(2+) channel mutations in APAs and primary aldosteronism.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Calcium Channels, L-Type/genetics , Germ-Line Mutation , Hyperaldosteronism/genetics , Mutation , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/biosynthesis , Amino Acid Sequence , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Cell Line , Child , Child, Preschool , Female , Humans , Hyperaldosteronism/metabolism , Male , Molecular Sequence Data , Pedigree , Protein Conformation , Sequence Alignment
20.
PLoS One ; 7(7): e41926, 2012.
Article in English | MEDLINE | ID: mdl-22848660

ABSTRACT

BACKGROUND: Aldosterone producing lesions are a common cause of hypertension, but genetic alterations for tumorigenesis have been unclear. Recently, either of two recurrent somatic missense mutations (G151R or L168R) was found in the potassium channel KCNJ5 gene in aldosterone producing adenomas. These mutations alter the channel selectivity filter and result in Na(+) conductance and cell depolarization, stimulating aldosterone production and cell proliferation. Because a similar mutation occurs in a mendelian form of primary aldosteronism, these mutations appear to be sufficient for cell proliferation and aldosterone production. The prevalence and spectrum of KCNJ5 mutations in different entities of adrenocortical lesions remain to be defined. MATERIALS AND METHODS: The coding region and flanking intronic segments of KCNJ5 were subjected to Sanger DNA sequencing in 351 aldosterone producing lesions, from patients with primary aldosteronism and 130 other adrenocortical lesions. The specimens had been collected from 10 different worldwide referral centers. RESULTS: G151R or L168R somatic mutations were identified in 47% of aldosterone producing adenomas, each with similar frequency. A previously unreported somatic mutation near the selectivity filter, E145Q, was observed twice. Somatic G151R or L168R mutations were also found in 40% of aldosterone producing adenomas associated with marked hyperplasia, but not in specimens with merely unilateral hyperplasia. Mutations were absent in 130 non-aldosterone secreting lesions. KCNJ5 mutations were overrepresented in aldosterone producing adenomas from female compared to male patients (63 vs. 24%). Males with KCNJ5 mutations were significantly younger than those without (45 vs. 54, respectively; p<0.005) and their APAs with KCNJ5 mutations were larger than those without (27.1 mm vs. 17.1 mm; p<0.005). DISCUSSION: Either of two somatic KCNJ5 mutations are highly prevalent and specific for aldosterone producing lesions. These findings provide new insight into the pathogenesis of primary aldosteronism.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Aldosterone/biosynthesis , Mutation , Sequence Analysis , Adolescent , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Adult , Aged , Base Sequence , Cohort Studies , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Mutation Rate , Sex Characteristics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL