Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Z Rheumatol ; 83(1): 54-67, 2024 Feb.
Article in German | MEDLINE | ID: mdl-38019334

ABSTRACT

The ability to visualize the nerves of the lower extremities differs from that of the upper extremities in sonography because the soft tissue cover is significantly larger in some cases. Landmarks are also defined for the lower extremities, which enable precise visualization of the nerves. Nerves and muscles are to be understood as a functional unit. In addition to the clarification of nerve compression syndromes, polyneuropathies and nerve tumors, sonography is also used to visualize muscle atrophy.


Subject(s)
Nerve Compression Syndromes , Polyneuropathies , Humans , Ultrasonography , Lower Extremity/diagnostic imaging
2.
J Immunother Cancer ; 11(11)2023 11.
Article in English | MEDLINE | ID: mdl-37963637

ABSTRACT

BACKGROUND: The metabolism of tryptophan to kynurenines (KYN) by indoleamine-2,3-dioxygenase or tryptophan-2,3-dioxygenase is a key pathway of constitutive and adaptive tumor immune resistance. The immunosuppressive effects of KYN in the tumor microenvironment are predominantly mediated by the aryl hydrocarbon receptor (AhR), a cytosolic transcription factor that broadly suppresses immune cell function. Inhibition of AhR thus offers an antitumor therapy opportunity via restoration of immune system functions. METHODS: The expression of AhR was evaluated in tissue microarrays of head and neck squamous cell carcinoma (HNSCC), non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). A structure class of inhibitors that block AhR activation by exogenous and endogenous ligands was identified, and further optimized, using a cellular screening cascade. The antagonistic properties of the selected AhR inhibitor candidate BAY 2416964 were determined using transactivation assays. Nuclear translocation, target engagement and the effect of BAY 2416964 on agonist-induced AhR activation were assessed in human and mouse cancer cells. The immunostimulatory properties on gene and cytokine expression were examined in human immune cell subsets. The in vivo efficacy of BAY 2416964 was tested in the syngeneic ovalbumin-expressing B16F10 melanoma model in mice. Coculture of human H1299 NSCLC cells, primary peripheral blood mononuclear cells and fibroblasts mimicking the human stromal-tumor microenvironment was used to assess the effects of AhR inhibition on human immune cells. Furthermore, tumor spheroids cocultured with tumor antigen-specific MART-1 T cells were used to study the antigen-specific cytotoxic T cell responses. The data were analyzed statistically using linear models. RESULTS: AhR expression was observed in tumor cells and tumor-infiltrating immune cells in HNSCC, NSCLC and CRC. BAY 2416964 potently and selectively inhibited AhR activation induced by either exogenous or endogenous AhR ligands. In vitro, BAY 2416964 restored immune cell function in human and mouse cells, and furthermore enhanced antigen-specific cytotoxic T cell responses and killing of tumor spheroids. In vivo, oral application with BAY 2416964 was well tolerated, induced a proinflammatory tumor microenvironment, and demonstrated antitumor efficacy in a syngeneic cancer model in mice. CONCLUSIONS: These findings identify AhR inhibition as a novel therapeutic approach to overcome immune resistance in various types of cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Dioxygenases , Head and Neck Neoplasms , Lung Neoplasms , Humans , Mice , Animals , Tryptophan , Receptors, Aryl Hydrocarbon/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Leukocytes, Mononuclear/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Lung Neoplasms/drug therapy , Kynurenine/metabolism , Immunotherapy , Immunologic Factors , Head and Neck Neoplasms/drug therapy , Tumor Microenvironment
4.
Mol Cell ; 77(6): 1322-1339.e11, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32006464

ABSTRACT

Deregulated expression of MYC induces a dependence on the NUAK1 kinase, but the molecular mechanisms underlying this dependence have not been fully clarified. Here, we show that NUAK1 is a predominantly nuclear protein that associates with a network of nuclear protein phosphatase 1 (PP1) interactors and that PNUTS, a nuclear regulatory subunit of PP1, is phosphorylated by NUAK1. Both NUAK1 and PNUTS associate with the splicing machinery. Inhibition of NUAK1 abolishes chromatin association of PNUTS, reduces spliceosome activity, and suppresses nascent RNA synthesis. Activation of MYC does not bypass the requirement for NUAK1 for spliceosome activity but significantly attenuates transcription inhibition. Consequently, NUAK1 inhibition in MYC-transformed cells induces global accumulation of RNAPII both at the pause site and at the first exon-intron boundary but does not increase mRNA synthesis. We suggest that NUAK1 inhibition in the presence of deregulated MYC traps non-productive RNAPII because of the absence of correctly assembled spliceosomes.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Protein Kinases/metabolism , Protein Phosphatase 1/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Spliceosomes/metabolism , Transcription, Genetic , Animals , Cell Nucleus/genetics , Chromatin/genetics , Gene Expression Regulation , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation , Protein Kinases/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Splicing , Repressor Proteins/genetics , Spliceosomes/genetics
5.
J Med Chem ; 63(2): 601-612, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31859507

ABSTRACT

The serine/threonine kinase TBK1 (TANK-binding kinase 1) and its homologue IKKε are noncanonical members of the inhibitor of the nuclear factor κB (IκB) kinase family. These kinases play important roles in multiple cellular pathways and, in particular, in inflammation. Herein, we describe our investigations on a family of benzimidazoles and the identification of the potent and highly selective TBK1/IKKε inhibitor BAY-985. BAY-985 inhibits the cellular phosphorylation of interferon regulatory factor 3 and displays antiproliferative efficacy in the melanoma cell line SK-MEL-2 but showed only weak antitumor activity in the SK-MEL-2 human melanoma xenograft model.


Subject(s)
I-kappa B Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Binding Sites , Crystallography, X-Ray , Drug Discovery , High-Throughput Screening Assays , Humans , Models, Molecular , Phosphorylation , Structure-Activity Relationship , Substrate Specificity
6.
J Med Chem ; 62(2): 928-940, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30563338

ABSTRACT

The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.


Subject(s)
Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Apoptosis/drug effects , Binding Sites , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , Half-Life , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyridines/metabolism , Pyridines/pharmacology , Pyrimidines/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription, Genetic/drug effects
7.
J Biol Chem ; 291(21): 11252-67, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27030009

ABSTRACT

The spindle assembly checkpoint (SAC) is an essential safeguarding mechanism devised to ensure equal chromosome distribution in daughter cells upon mitosis. The proteins Bub3 and BubR1 are key components of the mitotic checkpoint complex, an essential part of the molecular machinery on which the SAC relies. In the present work we have performed a detailed functional and biochemical characterization of the interaction between human Bub3 and BubR1 in cells and in vitro Our results demonstrate that genetic knockdown of Bub3 abrogates the SAC, promotes apoptosis, and inhibits the proliferation of human cancer cells. We also show that the integrity of the human mitotic checkpoint complex depends on the specific recognition between BubR1 and Bub3, for which the BubR1 Gle2 binding sequence motif is essential. This 1:1 binding event is high affinity, enthalpy-driven and with slow dissociation kinetics. The affinity, kinetics, and thermodynamic parameters of the interaction are differentially modulated by small regions in the N and C termini of the Gle2 binding domain sequence, suggesting the existence of "hotspots" for this protein-protein interaction. Furthermore, we show that specific disruption of endogenous BubR1·Bub3 complexes in human cancer cells phenocopies the effects observed in gene targeting experiments. Our work enhances the current understanding of key members of the SAC and paves the road for the pursuit of novel targeted cancer therapies based on SAC inhibition.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , M Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Apoptosis , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation , Gene Knockdown Techniques , HeLa Cells , Humans , Kinetics , M Phase Cell Cycle Checkpoints/genetics , MCF-7 Cells , Models, Molecular , Poly-ADP-Ribose Binding Proteins , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spindle Apparatus/genetics , Thermodynamics
8.
Exp Cell Res ; 323(1): 131-143, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24480576

ABSTRACT

Cancer cells in poorly vascularized tumor regions need to adapt to an unfavorable metabolic microenvironment. As distance from supplying blood vessels increases, oxygen and nutrient concentrations decrease and cancer cells react by stopping cell cycle progression and becoming dormant. As cytostatic drugs mainly target proliferating cells, cancer cell dormancy is considered as a major resistance mechanism to this class of anti-cancer drugs. Therefore, substances that target cancer cells in poorly vascularized tumor regions have the potential to enhance cytostatic-based chemotherapy of solid tumors. With three-dimensional growth conditions, multicellular tumor spheroids (MCTS) reproduce several parameters of the tumor microenvironment, including oxygen and nutrient gradients as well as the development of dormant tumor regions. We here report the setup of a 3D cell culture compatible high-content screening system and the identification of nine substances from two commercially available drug libraries that specifically target cells in inner MCTS core regions, while cells in outer MCTS regions or in 2D cell culture remain unaffected. We elucidated the mode of action of the identified compounds as inhibitors of the respiratory chain and show that induction of cell death in inner MCTS core regions critically depends on extracellular glucose concentrations. Finally, combinational treatment with cytostatics showed increased induction of cell death in MCTS. The data presented here shows for the first time a high-content based screening setup on 3D tumor spheroids for the identification of substances that specifically induce cell death in inner tumor spheroid core regions. This validates the approach to use 3D cell culture screening systems to identify substances that would not be detectable by 2D based screening in otherwise similar culture conditions.


Subject(s)
Antineoplastic Agents/isolation & purification , Enzyme Inhibitors/isolation & purification , Spheroids, Cellular/drug effects , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Electron Transport/drug effects , Enzyme Inhibitors/pharmacology , Female , Glucose/metabolism , Humans , Staurosporine/pharmacology , Tumor Cells, Cultured , Tumor Microenvironment/physiology
9.
Fertil Steril ; 99(3): 927-935.e6, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23260859

ABSTRACT

OBJECTIVE: To develop a predictive mouse model for uterine fibroids. DESIGN: Human fibroid cells xenografted to immunodeficient mice. SETTING: University and industrial research center. ANIMAL(S): Immunodeficient scid/beige mice. INTERVENTION(S): Subcutaneous and intrauterine injection of fibroid-derived cells, SV40 transformation of primary cells by lentiviral transduction, proliferation determined by immunohistochemistry, FISH. MAIN OUTCOME MEASURE(S): Characterization of primary and immortalized cells by Western blot and soft agar assay, determination of in vivo tumorigenicity, comparative histology and immunohistochemistry, fluorescence in situ hybridization. RESULT(S): Tumorigenicity of primary myoma cells disappears upon in vitro culture. Transformation and immortalization does not restore or conserve the in vivo growth potential of cultured cells. Injection of primary cells into myometrium of mice leads to xenografts with a leiomyoma-like histology. CONCLUSION(S): Primary myoma cells are suited to generate fibroid-like xenografts for studying pathogenesis without genetic modifications. In contrast, in vitro culture abolishes transplantability, and neither transformation nor immortalization is sufficient to restore tumorigenic capacity.


Subject(s)
Disease Models, Animal , Leiomyoma/pathology , Mice, SCID , Uterine Neoplasms/pathology , Adenocarcinoma/pathology , Animals , Cell Line, Transformed , Cell Line, Tumor , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Leiomyoma/physiopathology , Lung Neoplasms/pathology , Mice , Primary Cell Culture , Uterine Neoplasms/physiopathology , Xenograft Model Antitumor Assays
10.
PLoS One ; 7(5): e36125, 2012.
Article in English | MEDLINE | ID: mdl-22563479

ABSTRACT

Silencing of genes by hypermethylation contributes to cancer progression and has been shown to occur with increased frequency at specific genomic loci. However, the precise mechanisms underlying the establishment and maintenance of aberrant methylation marks are still elusive. The de novo DNA methyltransferase 3B (DNMT3B) has been suggested to play an important role in the generation of cancer-specific methylation patterns. Previous studies have shown that a reduction of DNMT3B protein levels induces antiproliferative effects in cancer cells that were attributed to the demethylation and reactivation of tumor suppressor genes. However, methylation changes have not been analyzed in detail yet. Using RNA interference we reduced DNMT3B protein levels in colon cancer cell lines. Our results confirm that depletion of DNMT3B specifically reduced the proliferation rate of DNMT3B-overexpressing colon cancer cell lines. However, genome-scale DNA methylation profiling failed to reveal methylation changes at putative DNMT3B target genes, even in the complete absence of DNMT3B. These results show that DNMT3B is dispensable for the maintenance of aberrant DNA methylation patterns in human colon cancer cells and they have important implications for the development of targeted DNA methyltransferase inhibitors as epigenetic cancer drugs.


Subject(s)
Cell Proliferation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , RNA Interference , Caco-2 Cells , Cell Line , Cell Line, Tumor , Cell Survival/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Immunoblotting , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , DNA Methyltransferase 3B
12.
Nat Commun ; 2: 395, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21772266

ABSTRACT

High attrition rates of novel anti-cancer drugs highlight the need for improved models to predict toxicity. Although polo-like kinase 1 (Plk1) inhibitors are attractive candidates for drug development, the role of Plk1 in primary cells remains widely unexplored. Therefore, we evaluated the utility of an RNA interference-based model to assess responses to an inducible knockdown (iKD) of Plk1 in adult mice. Here we show that Plk1 silencing can be achieved in several organs, although adverse events are rare. We compared responses in Plk1-iKD mice with those in primary cells kept under controlled culture conditions. In contrast to the addiction of many cancer cell lines to the non-oncogene Plk1, the primary cells' proliferation, spindle assembly and apoptosis exhibit only a low dependency on Plk1. Responses to Plk1-depletion, both in cultured primary cells and in our iKD-mouse model, correspond well and thus provide the basis for using validated iKD mice in predicting responses to therapeutic interventions.


Subject(s)
Antineoplastic Agents/toxicity , Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , RNA Interference/drug effects , Toxicity Tests/methods , Animals , Apoptosis/genetics , Blotting, Northern , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , DNA Primers/genetics , Drug Evaluation, Preclinical , Flow Cytometry , Fluorescent Antibody Technique , Gene Dosage/genetics , Gene Knockdown Techniques , Genetic Engineering/methods , Humans , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Polo-Like Kinase 1
13.
J Biol Chem ; 277(33): 29817-24, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12021264

ABSTRACT

Apoptotic cell death is of central importance in the pathogenesis of viral infections. Activation of a cascade of cysteine proteases, i.e. caspases, plays a key role in the effector phase of virus-induced apoptosis. However, little is known about pathways leading to the activation of initiator caspases in virus-infected host cells. Recently, we have shown that Sendai virus (SeV) infection triggers apoptotic cell death by activation of the effector caspase-3 and initiator caspase-8. We now investigated mechanisms leading to the activation of another initiator caspase, caspase-9. Unexpectedly we found that caspase-9 cleavage is not dependent on the presence of active caspases-3 or -8. Furthermore, the presence of caspase-9 in mouse embryonic fibroblast (MEF) cells was a prerequisite for Sendai virus-induced apoptotic cell death. Caspase-9 activation occurred without the release of cytochrome c from mitochondria and was not dependent on the presence of Apaf-1 or reactive oxygen intermediates. Our results therefore suggest an alternative mechanism for caspase-9 activation in virally infected cells beside the well characterized pathways via death receptors or mitochondrial cytochrome c release.


Subject(s)
Caspases/metabolism , Proteins/metabolism , Sendai virus/physiology , Animals , Apoptotic Protease-Activating Factor 1 , Caspase 8 , Caspase 9 , Enzyme Activation , Humans , Hydrolysis , Mice , Reactive Oxygen Species , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...