Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Oncogene ; 38(23): 4560-4573, 2019 06.
Article in English | MEDLINE | ID: mdl-30755730

ABSTRACT

Breast cancer is a heterogeneous genetic disease driven by the accumulation of individual mutations per tumor. Whole-genome sequencing approaches have identified numerous genes with recurrent mutations in primary tumors. Although mutations in well characterized tumor suppressors and oncogenes are overrepresented in these sets, the majority of the genetically altered genes have so far unknown roles in breast cancer progression. To improve the basic understanding of the complex disease breast cancer and to potentially identify novel drug targets or regulators of known cancer-driving pathways, we analyzed 86 wild-type genes and 94 mutated variants for their effect on cell growth using a serially constructed panel of MCF7 cell lines. We demonstrate in subsequent experiments that the metal cation transporter CNNM4 regulates growth by induction of apoptosis and identified a tumor suppressive role of complement factor properdin (CFP) in vitro and in vivo. CFP appears to induce the intracellular upregulation of the pro-apoptotic transcription factor DDIT3 which is associated with endoplasmic reticulum-stress response.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , LIM Domain Proteins/metabolism , Properdin/metabolism , Transcription Factor CHOP/metabolism , Animals , Apoptosis , Cations , Cell Proliferation/drug effects , Cell Survival , Disease Progression , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Transplantation , Phenotype , RNA-Binding Proteins , Sequence Analysis, DNA , Up-Regulation
2.
PLoS One ; 13(1): e0189887, 2018.
Article in English | MEDLINE | ID: mdl-29293529

ABSTRACT

Cancer results from alterations at essential genomic sites and is characterized by uncontrolled cell proliferation, invasion and metastasis. Identification of driver genes of metastatic progression is essential, as metastases, not primary tumors, are fatal. To gain insight into the mutational concordance between different steps of malignant progression we performed exome sequencing and validation with targeted deep sequencing of successive steps of malignant progression from pre-invasive stages to asynchronous distant metastases in six breast cancer patients. Using the ratio of non-synonymous to synonymous mutations, a surprisingly large number of cancer driver genes, ranging between 3 and 145, were estimated to confer a selective advantage in the studied primary tumors. We report a substantial amount of metastasis specific mutations and a number of novel putative metastasis driver genes. Most notable are the DCC, ABCA13, TIAM2, CREBBP, BCL6B and ZNF185 genes, mainly mutated exclusively in metastases and highly likely driver genes of metastatic progression. We find different genes and pathways to be affected at different steps of malignant progression. The Adherens junction pathway is affected in four of the six studied patients and this pathway most likely plays a vital role in the metastatic process.


Subject(s)
Breast Neoplasms/pathology , High-Throughput Nucleotide Sequencing/methods , Neoplasm Metastasis/genetics , Breast Neoplasms/genetics , Female , Humans
3.
Anal Biochem ; 542: 16-19, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29169777

ABSTRACT

Deleted in Malignant Brain Tumor 1 (DMBT1, alias SAG or gp340) is a pattern recognition receptor involved in immune defense, cell polarization, differentiation and regeneration. To investigate the role of the protein in physiological and pathological processes, the protein has often been isolated from saliva or produced in vitro and purified by a multistep affinity purification procedure using bacteria, followed by FPLC. Here, we compared a simple, one-step FPLC-SEC protocol for purification of recombinant DMBT1 6 kb, with that of the standard bacteria affinity purification-based protocol. Our data suggest that our FPLC-SEC protocol yields DMBT1 in a more native conformation.


Subject(s)
Receptors, Cell Surface/isolation & purification , Receptors, Cell Surface/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Chromatography, Gel , Chromatography, Liquid , DNA-Binding Proteins , Humans , Receptors, Cell Surface/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Streptococcus mutans/chemistry , Streptococcus mutans/metabolism , Tumor Suppressor Proteins
4.
Mol Ther Nucleic Acids ; 8: 264-276, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918028

ABSTRACT

Small interfering RNA (siRNA) is a promising molecule for gene therapy, but its therapeutic administration remains problematic. Among the recently proposed vectors, cell-penetrating peptides show great promise in in vivo trials for siRNA delivery. Human protein DMBT1 (deleted in malignant brain tumor 1) is a pattern recognition molecule that interacts with polyanions and recognizes and aggregates bacteria. Taking advantage of these properties, we investigated whether specific synthetic DMBT1-derived peptides could be used to formulate nanoparticles for siRNA administration. Using an electrophoretic mobility shift assay and UV spectra, we identified two DMBT1 peptides that could encapsulate the siRNA with a self- and co-assembly mechanism. The complexes were stable for at least 2 hr in the presence of either fetal bovine serum (FBS) or RNase A, with peptide-dependent time span protection. ζ-potential, circular dichroism, dynamic light scattering, and transmission electron microscopy revealed negatively charged nanoparticles with an average diameter of 10-800 nm, depending on the reaction conditions, and a spherical or rice-shaped morphology, depending on the peptide and ß-helix conformation. We successfully transfected human MCF7 cells with fluorescein isothiocyanate (FITC)-DMBT1-peptide-Cy3-siRNA complexes. Finally, DMBT1 peptides encapsulating an siRNA targeting a fluorescent reporter gene showed efficient gene silencing in MCF7-recombinant cells. These results lay the foundation for a new research line to exploit DMBT1-peptide nanocomplexes for therapeutic siRNA delivery.

6.
PLoS One ; 12(5): e0178286, 2017.
Article in English | MEDLINE | ID: mdl-28542563

ABSTRACT

Cancer stem cells represent the putative tumor-driving subpopulation thought to account for drug resistance, relapse, and metastatic spread of epithelial and other cancer types. Accordingly, cell surface markers for therapeutic delivery to cancer stem cells are subject of intense research. Somatostatin receptor 2 and nucleolin are known to be overexpressed by various cancer types, which have elicited comprehensive efforts to explore their therapeutic utilization. Here, we evaluated somatostatin receptor 2 targeting and nucleolin targeting for therapeutic delivery to cancer stem cells from lung cancer. Nucleolin is expressed highly but not selectively, while somatostatin receptor 2 is expressed selectively but not highly by cancer cells. The non-small cell lung cancer cell lines A549 and H1299, displayed average levels of both surface molecules as judged based on analysis of a larger cell line panel. H1299 compared to A549 cells showed significantly elevated sphere-forming capacity, indicating higher cancer stem cell content, thus qualifying as suitable test system. Nucleolin-targeting 57Co-DOTA-AS1411 aptamer showed efficient internalization by cancer cells and, remarkably, at even higher efficiency by cancer stem cells. In contrast, somatostatin receptor 2 expression levels were not sufficiently high in H1299 cells to confer efficient uptake by either non-cancer stem cells or cancer stem cells. The data provides indication that the nucleolin-targeting AS1411 aptamer might be used for therapeutic delivery to non-small cell lung cancer stem cells.


Subject(s)
Aptamers, Nucleotide/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Heterocyclic Compounds, 1-Ring/chemistry , Neoplastic Stem Cells/drug effects , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Drug Delivery Systems , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Somatostatin/pharmacology , Tumor Cells, Cultured , Nucleolin
7.
Immunogenetics ; 69(6): 401-407, 2017 06.
Article in English | MEDLINE | ID: mdl-28364129

ABSTRACT

The Scavenger Receptor Cysteine-Rich (SRCR) proteins are an archaic group of proteins characterized by the presence of multiple SRCR domains. They are membrane-bound or secreted proteins, which are generally related to host defense systems in animals. Deleted in Malignant Brain Tumors 1 (DMBT1) is a SRCR protein which is secreted in mucosal fluids and involved in host defense by pathogen binding by its SRCR domains. Genetic polymorphism within DMBT1 leads to DMBT1-alleles giving rise to polypeptides with interindividually different numbers of SRCR domains, ranging from 8 SRCR domains (encoded by 6 kb DMBT1 variant) to 13 SRCR domains (encoded by the 8 kb DMBT1 variant). In the present study, we have investigated whether reduction from 13 to 8 amino-terminal SRCR domains leads to reduction of bacterial binding. The 6 kb variant bound ~20-45% less bacteria compared to the 8 kb variant. These results support the hypothesis that genetic variation in DMBT1 may influence microbial defense.


Subject(s)
Germ-Line Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Sequence Deletion , Bacterial Adhesion/genetics , Calcium-Binding Proteins , DNA-Binding Proteins , Humans , Polymorphism, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Cell Surface/chemistry , Receptors, Scavenger/chemistry , Tumor Suppressor Proteins
8.
Oncotarget ; 8(14): 23760-23774, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28423600

ABSTRACT

Next-generation sequencing has dramatically increased genome-wide profiling options and conceptually initiates the possibility for personalized cancer therapy. State-of-the-art sequencing studies yield large candidate gene sets comprising dozens or hundreds of mutated genes. However, few technologies are available for the systematic downstream evaluation of these results to identify novel starting points of future cancer therapies.We improved and extended a site-specific recombination-based system for systematic analysis of the individual functions of a large number of candidate genes. This was facilitated by a novel system for the construction of isogenic constitutive and inducible gain- and loss-of-function cell lines. Additionally, we demonstrate the construction of isogenic cell lines with combinations of the traits for advanced functional in vitro analyses. In a proof-of-concept experiment, a library of 108 isogenic melanoma cell lines was constructed and 8 genes were identified that significantly reduced viability in a discovery screen and in an independent validation screen. Here, we demonstrate the broad applicability of this recombination-based method and we proved its potential to identify new drug targets via the identification of the tumor suppressor DUSP6 as potential synthetic lethal target in melanoma cell lines with BRAF V600E mutations and high DUSP6 expression.


Subject(s)
Dual Specificity Phosphatase 6/genetics , Melanoma/genetics , Cell Line, Tumor , Dual Specificity Phosphatase 6/biosynthesis , Dual Specificity Phosphatase 6/metabolism , Gene Knockdown Techniques , Humans , Mass Screening , Melanoma/metabolism , Melanoma/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Recombination, Genetic , Transfection
9.
Sci Rep ; 7: 43813, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276460

ABSTRACT

A main controversy in cancer research is whether metastatic abilities are present in the most advanced clone of the primary tumor or result from independently acquired aberrations in early disseminated cancer cells as suggested by the linear and the parallel progression models, respectively. The genetic concordance between different steps of malignant progression is mostly unexplored as very few studies have included cancer samples separated by both space and time. We applied whole exome sequencing and targeted deep sequencing to 26 successive samples from six patients with metastatic estrogen receptor (ER)-positive breast cancer. Our data provide support for both linear and parallel progression towards metastasis. We report for the first time evidence of metastasis-to-metastasis seeding in breast cancer. Our results point to three distinct routes of metastasis emergence. This may have profound clinical implications and provides substantial novel molecular insights into the timing and mutational evolution of breast cancer metastasis.


Subject(s)
Bone Neoplasms/genetics , Breast Neoplasms/genetics , Liver Neoplasms/genetics , Mutation , Aged , Aged, 80 and over , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cytoskeletal Proteins/genetics , Disease Progression , Female , Genomics , Humans , LIM Domain Proteins/genetics , Liver Neoplasms/secondary , Middle Aged , Repressor Proteins/genetics , Exome Sequencing/methods
10.
J Neurooncol ; 132(1): 45-54, 2017 03.
Article in English | MEDLINE | ID: mdl-28091986

ABSTRACT

Glioblastoma multiforme (GBM) is the most frequent malignant primary brain tumor. A major reason for the overall median survival being only 14.6 months is migrating tumor cells left behind after surgery. Another major reason is tumor cells having a so-called cancer stem cell phenotype being therefore resistant towards traditional chemo- and radiotherapy. A group of novel molecular targets are microRNAs (miRNAs). MiRNAs are small non-coding RNAs exerting post-transcriptional regulation of gene expression. The aim of this study was to identify differentially expressed miRNAs in migrating GBM cells using serum-free stem cell conditions. We used patient-derived GBM spheroid cultures for a novel serum-free migration assay. MiRNA expression of migrating tumor cells isolated at maximum migration speed was compared with corresponding spheroids using an OpenArray Real-Time PCR System. The miRNA profiling revealed 30 miRNAs to be differentially expressed. In total 13 miRNAs were upregulated and 17 downregulated in migrating cells compared to corresponding spheroids. The three most deregulated miRNAs, miR-1227 (up-regulated), miR-32 (down-regulated) and miR-222 (down-regulated), were experimentally overexpressed. A non-significantly increased migration rate was observed after miR-1227 overexpression. A significantly reduced migration rate was observed after miR-32 and miR-222 overexpression. In conclusion a shift in microRNA profile upon glioma cell migration was identified using an assay avoiding serum-induced migration. Both the miRNA profiling and the functional validation suggested that miR-1227 may be associated with increased migration and miR-32 and miR-222 with decreased migration. These miRNAs may represent potential novel targets in migrating glioma cells.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement , Glioblastoma/metabolism , MicroRNAs/metabolism , Culture Media, Serum-Free , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Spheroids, Cellular , Tumor Cells, Cultured
11.
Nanomedicine ; 13(3): 1229-1233, 2017 04.
Article in English | MEDLINE | ID: mdl-28064007

ABSTRACT

The nanomaterial community calls for standardized in vitro assays to determine nanoparticle toxicity in the effort to reduce the number of in vivo validation experiments. We demonstrate that chip-based protein detection is suitable for assessing toxicity and may complement traditional assays to improve selection of primary hits for subsequent analysis. As nanodrug mimics, we analyzed the effect of transiently transfected siRNAs in MCF7 breast cancer cells and normal MCF12A breast cells, resembling a differential screen. As a measure of cytotoxicity, we determined cell viability as well as protein expression of glyceraldehyde-3-phosphate dehydrogenase, transferrin receptor, and the proliferation marker Ki67. The evaluation of cell lethality and protein expression unraveled cellular effects overseen by one method alone.


Subject(s)
Nanoparticles/toxicity , Protein Array Analysis/methods , Toxicity Tests/methods , Breast/cytology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Expression Regulation/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Ki-67 Antigen/genetics , RNA, Small Interfering/genetics , Receptors, Transferrin/genetics , Transfection
12.
Histopathology ; 70(7): 1064-1071, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28130841

ABSTRACT

AIMS: Deleted in malignant brain tumours 1 (DMBT1) exerts functions in the regulation of epithelial differentiation and inflammation and has been proposed as a tumour suppressor. Because chronic inflammation is a hallmark of cholangiocarcinogenesis, the aim of this study was to investigate the expression of DMBT1 in biliary tract cancer (BTC) and to correlate this expression with clinicopathological data. METHODS AND RESULTS: The expression of DMBT1 protein was examined immunohistochemically in 157 BTC patients [41 intrahepatic (ICC), 60 extrahepatic cholangiocarcinomas (ECC) and 56 adenocarcinomas of the gallbladder (GBAC)]. Additionally, 56 samples of high-grade biliary intraepithelial neoplasia (BilIN 3) and 92 corresponding samples of histological non-neoplastic biliary tract tissues were included. DMBT1 expression was increased significantly in BilIN 3 compared to normal tissue (P < 0.0001) and BTC (P < 0.0001). BTC showed no significant difference in DMBT1 expression compared to non-neoplastic biliary tissue (P = 0.315). Absent DMBT1 expression in non-neoplastic biliary tissue of BTC patients was associated with poorer survival (P = 0.027). DMBT1 expression was correlated significantly with patients' age (P < 0.001). CONCLUSION: DMBT1 is expressed differently in cholangiocarcinogenesis and poorer patients' survival rates are associated with absent DMBT1 expression in non-neoplastic biliary tissue, suggesting a tumour-suppressive role of DMBT1 in early cholangiocarcinogenesis.


Subject(s)
Bile Duct Neoplasms/pathology , Biliary Tract Neoplasms/pathology , Biomarkers, Tumor/analysis , Receptors, Cell Surface/biosynthesis , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/mortality , Biliary Tract Neoplasms/metabolism , Biliary Tract Neoplasms/mortality , Calcium-Binding Proteins , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Cohort Studies , DNA-Binding Proteins , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models , Receptors, Cell Surface/analysis , Tumor Suppressor Proteins
13.
SLAS Discov ; 22(2): 196-202, 2017 02.
Article in English | MEDLINE | ID: mdl-27729504

ABSTRACT

High-throughput screening (HTS) has become an indispensable tool for the pharmaceutical industry and for biomedical research. A high degree of automation allows for experiments in the range of a few hundred up to several hundred thousand to be performed in close succession. The basis for such screens are molecular libraries, that is, microtiter plates with solubilized reagents such as siRNAs, shRNAs, miRNA inhibitors or mimics, and sgRNAs, or small compounds, that is, drugs. These reagents are typically condensed to provide enough material for covering several screens. Library plates thus need to be serially diluted before they can be used as assay plates. This process, however, leads to an explosion in the number of plates and samples to be tracked. Here, we present SAVANAH, the first tool to effectively manage molecular screening libraries across dilution series. It conveniently links (connects) sample information from the library to experimental results from the assay plates. All results can be exported to the R statistical environment or piped into HiTSeekR ( http://hitseekr.compbio.sdu.dk ) for comprehensive follow-up analyses. In summary, SAVANAH supports the HTS community in managing and analyzing HTS experiments with an emphasis on serially diluted molecular libraries.


Subject(s)
High-Throughput Screening Assays/methods , MicroRNAs/antagonists & inhibitors , RNA, Small Interfering/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Computational Biology/methods , Humans , MicroRNAs/chemistry , Quality Control , RNA Interference , RNA, Small Interfering/chemistry , Small Molecule Libraries/chemistry , Software
14.
Histochem Cell Biol ; 147(3): 389-397, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27628193

ABSTRACT

Meconium aspiration syndrome (MAS) is characterized by surfactant inactivation and inflammation. As lung epithelial cells up-regulate nitric oxide (NO) in response to inflammation, the NO production following meconium exposition was examined in relation to expression of Deleted in Malignant Brain Tumors 1 (DMBT1), a protein with functions in innate immunity and inflammatory regulation. Here, DMBT1 expression was analyzed by immunohistochemistry in postmortem lung sections from patients with MAS. The lung epithelial cell line A549, stably transfected with a DMBT1 (DMBT1+ cells) expression plasmid or with an empty expression plasmid (DMBT1- cells), was exposed to meconium. NO was determined in dependence of aminoguanidine (inducible NO synthase inhibitor), steroids and lipopolysaccharide (LPS). DMBT1 is highly expressed in lungs with MAS. In the absence of meconium, DMBT1+ cells showed a higher NO production than the DMBT1- cells (p = 0.0090). Meconium led in DMBT1- and DMBT1+ cells to elevated NO levels (p < 0.0001), but with a higher NO level in DMBT1+ cells (p < 0.0001). Aminoguanidine, an iNOS inhibitor, reduced the higher NO production in DMBT1+ cells (p = 0.0476), but NO levels remained above NO production from DMBT1- cells (p = 0.0289). Dexamethasone diminished NO production in DMBT1+ cells after meconium exposition (p = 0.0076). Combined addition of LPS and meconium significantly increased NO production in both cell types (p < 0.0001). In comparison to exposure with only meconium, the combined addition of LPS and meconium to the cells increased NO levels in both DMBT1- cells (p = 0.0030) and DMBT1+ cells (p = 0.0028). In conclusion, basal and meconium-induced NO production in lung epithelial cells is positively regulated by DMBT1.


Subject(s)
Epithelial Cells/metabolism , Lung/cytology , Meconium/metabolism , Nitric Oxide/biosynthesis , Receptors, Cell Surface/metabolism , Calcium-Binding Proteins , DNA-Binding Proteins , Epithelial Cells/cytology , Humans , Immunohistochemistry , Infant, Newborn , Receptors, Cell Surface/chemistry , Tumor Suppressor Proteins
15.
J Drug Target ; 24(9): 836-856, 2016 11.
Article in English | MEDLINE | ID: mdl-27646195

ABSTRACT

The story starts in Basel at CLINAM in 2013, when I asked Pieter about making nanoparticles and he advised me to "try this solvent-exchange method we have developed for making limit sized particles". We are particularly interested in what are "limit size materials" because we want to test the feasibility of an idea: could we design, make, develop, and test the concept for treating metastatic cancer by, "Putting the Drug in the Cancer's Food? "Limit size" is the size of the cancer's food, ? the common Low Density Lipoprotein, (LDL) ~20 nm diameter. In this contribution to Pieter's LTAA we focus on the "bottom" (nucleation) and the "up" (growth) of "bottom-up design" as it applies to homogeneous nucleation of especially, hydrophobic drugs and the 8 physico-chemical stages and associated parameters that determine the initial size, and any subsequent coarsening, of a nanoparticle suspension. We show that, when made by the rapid solvent-exchange method, the same sized particles can be obtained without phospholipid. Furthermore, the obtained size follows the predictions of classic nucleation theory when the appropriate values for the parameters (surface tension and supersaturation) at nucleation are included. Calculations on dissolution time for nanoparticles reveal that a typical fewmicromolar-solubility, hydrophobic, anti-cancer drug (like Lapatinib, Niclosamide, Abiraterone, and Fulvestrant) of 500 nm diameter would take between 3?7 s to dissolve in an infinite sink like the blood stream; and a 50 nm particle would dissolve in less than a second! And so the nanoparticle design requires a highly water-insoluble drug, and a tight, encapsulating, impermeable lipid:cholesterol monolayer. While the "Y" junction can be used to mix an ethanolic solution with anti-solvent, we find that a "no-junction" can give equally good results. A series of nanoparticles (DiI-fluorescently labeled Triolein-cored and drug-cored nanoparticles of Orlistat) were then tested in well-characterized cell lines for uptake and efficacy as well as a PET-imageable nanoparticle in initial PET-imaging studies in animals for EPR uptake and tumor detection. We show that, while free-drug cannot be optimally administered in vivo, a nanoparticle formulation of orlistat could in principle represent a stable parenteral delivery system. The article ends with a brief discussion of what we see as the way forward in Individualized Medicine from the Diagnostic-Therapeutic ("Diapeutic") side, requiring 18FDG detection of metastatic lesions, functional imaging of a protein target (e.g. Fatty Acid Synthase) using 11C acetate, then a PET (or other)-imageable nanoparticle to demonstrate EPR accumulation, and then the administration of the pure-drug nanoparticle taken in by the most aggressive cancer cells in the perivascular space, as they would their "food".


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Nanoparticles , Neoplasms/drug therapy , Animals , Chemistry, Pharmaceutical , Humans , Hydrophobic and Hydrophilic Interactions , Lipids/chemistry
16.
F1000Res ; 5: 1531, 2016.
Article in English | MEDLINE | ID: mdl-27540470

ABSTRACT

Identifying functional modules or novel active pathways, recently termed de novo pathway enrichment, is a computational systems biology challenge that has gained much attention during the last decade. Given a large biological interaction network, KeyPathwayMiner extracts connected subnetworks that are enriched for differentially active entities from a series of molecular profiles encoded as binary indicator matrices. Since interaction networks constantly evolve, an important question is how robust the extracted results are when the network is modified. We enable users to study this effect through several network perturbation techniques and over a range of perturbation degrees. In addition, users may now provide a gold-standard set to determine how enriched extracted pathways are with relevant genes compared to randomized versions of the original network.

17.
Nucleic Acids Res ; 44(14): 6639-48, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27330136

ABSTRACT

High-throughput screening (HTS) is an indispensable tool for drug (target) discovery that currently lacks user-friendly software tools for the robust identification of putative hits from HTS experiments and for the interpretation of these findings in the context of systems biology. We developed HiTSeekR as a one-stop solution for chemical compound screens, siRNA knock-down and CRISPR/Cas9 knock-out screens, as well as microRNA inhibitor and -mimics screens. We chose three use cases that demonstrate the potential of HiTSeekR to fully exploit HTS screening data in quite heterogeneous contexts to generate novel hypotheses for follow-up experiments: (i) a genome-wide RNAi screen to uncover modulators of TNFα, (ii) a combined siRNA and miRNA mimics screen on vorinostat resistance and (iii) a small compound screen on KRAS synthetic lethality. HiTSeekR is publicly available at http://hitseekr.compbio.sdu.dk It is the first approach to close the gap between raw data processing, network enrichment and wet lab target generation for various HTS screen types.


Subject(s)
Drug Evaluation, Preclinical , High-Throughput Screening Assays/methods , Caspases/metabolism , Drug Delivery Systems , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Quality Control , RNA Interference , Robotics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
18.
Nucleic Acids Res ; 44(W1): W98-W104, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27150809

ABSTRACT

We present KeyPathwayMinerWeb, the first online platform for de novo pathway enrichment analysis directly in the browser. Given a biological interaction network (e.g. protein-protein interactions) and a series of molecular profiles derived from one or multiple OMICS studies (gene expression, for instance), KeyPathwayMiner extracts connected sub-networks containing a high number of active or differentially regulated genes (proteins, metabolites) in the molecular profiles. The web interface at (http://keypathwayminer.compbio.sdu.dk) implements all core functionalities of the KeyPathwayMiner tool set such as data integration, input of background knowledge, batch runs for parameter optimization and visualization of extracted pathways. In addition to an intuitive web interface, we also implemented a RESTful API that now enables other online developers to integrate network enrichment as a web service into their own platforms.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , Huntingtin Protein/genetics , Huntington Disease/genetics , User-Computer Interface , Case-Control Studies , Computational Biology/statistics & numerical data , Datasets as Topic , Gene Expression Profiling , Gene Expression Regulation , Humans , Huntington Disease/diagnosis , Internet , Protein Interaction Mapping
19.
J Neurooncol ; 128(3): 395-404, 2016 07.
Article in English | MEDLINE | ID: mdl-27063952

ABSTRACT

Glioblastomas always recur despite surgery, radiotherapy and chemotherapy. A key player in the therapeutic resistance may be immature tumor cells with stem-like properties (TSCs) escaping conventional treatment. A group of promising molecular targets are microRNAs (miRs). miRs are small non-coding RNAs exerting post-transcriptional regulation of gene expression. In this study we aimed to identify over-expressed TSC-related miRs potentially amenable for therapeutic targeting. We used non-differentiated glioblastoma spheroid cultures (GSCs) containing TSCs and compared these to xenografts using a NanoString nCounter platform. This revealed 19 over-expressed miRs in the non-differentiated GSCs. Additionally, non-differentiated GSCs were compared to neural stem cells (NSCs) using a microarray platform. This revealed four significantly over-expressed miRs in the non-differentiated GSCs in comparison to the NSCs. The three most over-expressed miRs in the non-differentiated GSCs compared to xenografts were miR-126, -137 and -128. KEGG pathway analysis suggested the main biological function of these over-expressed miRs to be cell-cycle arrest and diminished proliferation. To functionally validate the profiling results suggesting association of these miRs with stem-like properties, experimental over-expression of miR-128 was performed. A consecutive limiting dilution assay confirmed a significantly elevated spheroid formation in the miR-128 over-expressing cells. This may provide potential therapeutic targets for anti-miRs to identify novel treatment options for GBM patients.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Heterografts , Humans , Male , Microarray Analysis , Neoplasm Transplantation , Neural Stem Cells/metabolism , Rats, Nude , Spheroids, Cellular/transplantation
20.
Histochem Cell Biol ; 145(2): 227-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26542257

ABSTRACT

Deleted in malignant brain tumor 1 (DMBT1) is involved in innate immunity and epithelial differentiation. Previous studies in adults indicated a strong intestinal expression of DMBT1 and an important role in inflammatory bowel diseases. Here, we analyzed the DMBT1 expression in the fetal gastrointestinal system depending on gestational age and in patients with necrotizing enterocolitis (NEC), volvulus, intestinal perforation (IP), or herniation, representing typical diseases of preterm and term infants. We used immunohistochemistry and RNA in situ hybridization to detect DMBT1 protein and mRNA in fetal tissues, supplemented by postmortem analysis of DMBT1 expression in died newborns and analysis of surgically removed tissues. DMBT1 expression is detectable in the early developmental stages of the gastrointestinal system. In NEC, volvulus, IP, or herniation, characterized by high systemic inflammatory responses, DMBT1 expression is strongly increased. High DMBT1 expression was also found in the bile ducts of older infants with sepsis or cholestasis. The study shows that DMBT1 expression is observed in the developing gastrointestinal system and up-regulated in infants with NEC, volvulus, IP, and herniation. DMBT1 may play a role in epithelial differentiation and local innate immunity during neonatal inflammatory bowel processes.


Subject(s)
Gastrointestinal Diseases/metabolism , Receptors, Cell Surface/analysis , Receptors, Cell Surface/metabolism , Calcium-Binding Proteins , DNA-Binding Proteins , Gastrointestinal Diseases/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Receptors, Cell Surface/biosynthesis , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL