Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Chembiochem ; 25(13): e202400024, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38716781

ABSTRACT

Lagunamide A is a biologically active natural product with a yet unidentified molecular mode of action. Cellular studies revealed that lagunamide A is a potent inhibitor of cancer cell proliferation, promotes apoptosis and causes mitochondrial dysfunction. To decipher the cellular mechanism responsible for these effects, we utilized thermal protein profiling (TPP) and identified EYA3 as a stabilized protein in cells upon lagunamide A treatment. EYA3, involved in the DNA damage repair process, was functionally investigated via siRNA based knockdown studies and corresponding effects of lagunamide A on DNA repair were confirmed. Furthermore, we showed that lagunamide A sensitized tumor cells to treatment with the drug doxorubicin highlighting a putative therapeutic strategy.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , DNA Damage , DNA Repair , Proteome , Humans , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Proteome/drug effects , Proteome/metabolism , Proteome/analysis , Cell Line, Tumor , Doxorubicin/pharmacology
2.
Biol Chem ; 403(4): 421-431, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35224953

ABSTRACT

Since the first report on a yeast three-hybrid system, several approaches have successfully utilized different setups for discovering targets of small molecule drugs. Compared to broadly applied MS based target identification approaches, the yeast three-hybrid system represents a complementary method that allows for the straightforward identification of direct protein binders of selected small molecules. One major drawback of this system, however, is that the drug has to be taken up by the yeast cells in sufficient concentrations. Here, we report the establishment of a yeast three-hybrid screen in the deletion strain ABC9Δ, which is characterized by being highly permeable to small molecules. We used this system to screen for protein binding partners of ethinylestradiol, a widely used drug mainly for contraception and hormone replacement therapy. We identified procollagen-lysine 2-oxoglutarate 5-dioxygenase 2 (PLOD2 or lysyl hydroxylase, LH2) as a novel direct target and were able to confirm the interaction identified with the yeast three-hybrid system by a complementary method, affinity chromatography, to prove the validity of the hit. Furthermore, we provide evidence for an interaction between the drug and PLOD2 in vitro and in cellulo.


Subject(s)
Ethinyl Estradiol , Saccharomyces cerevisiae , Ethinyl Estradiol/pharmacology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Protein Binding , Saccharomyces cerevisiae/metabolism , Two-Hybrid System Techniques
3.
Angew Chem Int Ed Engl ; 59(4): 1595-1600, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31658409

ABSTRACT

Novel targets are needed for treatment of devastating diseases such as cancer. For decades, natural products have guided innovative therapies by addressing diverse pathways. Inspired by the potent cytotoxic bioactivity of myxobacterial vioprolides A-D, we performed in-depth studies on their mode of action. Based on its prominent potency against human acute lymphoblastic leukemia (ALL) cells, we conducted thermal proteome profiling (TPP) and deciphered the target proteins of the most active derivative vioprolide A (VioA) in Jurkat cells. Nucleolar protein 14 (NOP14), which is essential in ribosome biogenesis, was confirmed as a specific target of VioA by a suite of proteomic and biological follow-up experiments. Given its activity against ALL cells compared to healthy lymphocytes, VioA exhibits unique therapeutic potential for anticancer therapy through a novel mode of action.


Subject(s)
Biological Products/chemistry , Nuclear Proteins/chemistry , Humans , Ribosomes/metabolism
4.
ACS Cent Sci ; 5(7): 1170-1178, 2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31403069

ABSTRACT

The natural product neocarzilin A (NCA) was discovered decades ago, and despite its potent cytotoxic effects no mode of action studies have been performed up to date. Synthesis of neocarzilins A, B, and C and a stereoisomer of NCA provided insights into structural preferences as well as access to probes for functional studies. NCA turned out to be the most active member and was not only effective against cell proliferation but also migration, a novel and so far overlooked activity. To decipher the molecular mode of action, we applied chemical proteomics for target discovery and revealed that NCA targets cancer cell migration via irreversible binding to the largely uncharacterized synaptic vesicle membrane protein VAT-1. A corresponding knockout of the protein confirmed the phenotype, and pull-down studies showed the interaction with an intricate network of key migration mediators such as Talin-1. Overall, we introduce VAT-1 as a promising novel target for the development of selective migration inhibitors with the perspective to limit toxicity in the absence of antiproliferative effects.

5.
Oncogene ; 38(5): 609-621, 2019 01.
Article in English | MEDLINE | ID: mdl-30171260

ABSTRACT

Melanoma is the deadliest form of skin cancer with rising incidence, creating a significant health problem. We discovered increased expression of bone morphogenetic protein 6 (BMP6) in melanoma cells and tissues, and observed that BMP6 deficiency caused significantly delayed tumor onset and decelerated tumor progression in a melanoma mouse model. Moreover, we determined that BMP6 inhibits dermal mast cell recruitment and found that mast cell-derived mediators significantly reduced melanoma growth in vitro. In line with this, mast cell deficiency accelerated tumor onset and progression in a melanoma mouse model. Analysis of human melanoma tissues revealed a strong negative correlation between melanoma proliferation and mast cell infiltration. This study elucidates a novel role of BMP6-induced modulation of the tumor microenvironment.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Skin Neoplasms/metabolism , Tumor Microenvironment , Animals , Bone Morphogenetic Protein 6/genetics , Cell Line, Tumor , Female , Humans , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology
6.
Haematologica ; 104(3): 546-555, 2019 03.
Article in English | MEDLINE | ID: mdl-30309851

ABSTRACT

Combination chemotherapy has proven to be a favorable strategy to treat acute leukemia. However, the introduction of novel compounds remains challenging and is hindered by a lack of understanding of their mechanistic interactions with established drugs. In the present study, we demonstrate a highly increased response of various acute leukemia cell lines, drug-resistant cells and patient-derived xenograft cells by combining the recently introduced protein disulfide isomerase inhibitor PS89 with cytostatics. In leukemic cells, a proteomics-based target fishing approach revealed that PS89 affects a whole network of endoplasmic reticulum homeostasis proteins. We elucidate that the strong induction of apoptosis in combination with cytostatics is orchestrated by the PS89 target B-cell receptor-associated protein 31, which transduces apoptosis signals at the endoplasmic reticulum -mitochondria interface. Activation of caspase-8 and cleavage of B-cell receptor-associated protein 31 stimulate a pro-apoptotic crosstalk including release of calcium from the endoplasmic reticulum and an increase in the levels of reactive oxygen species resulting in amplification of mitochondrial apoptosis. The findings of this study promote PS89 as a novel chemosensitizing agent for the treatment of acute leukemia and uncovers that targeting the endoplasmic reticulum - mitochondrial network of cell death is a promising approach in combination therapy.


Subject(s)
Cytostatic Agents/pharmacology , Endoplasmic Reticulum/metabolism , Leukemia/metabolism , Mitochondria/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Humans , Leukemia/drug therapy , Leukemia/pathology , Mice , Models, Biological , Proteome , Proteomics/methods , Xenograft Model Antitumor Assays
7.
Biotechnol Adv ; 36(6): 1724-1729, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29447918

ABSTRACT

Since cancer cells depend on de novo lipogenesis for energy supply, highly active membrane biosynthesis and signaling, enhanced fatty acid synthesis is a crucial characteristic of cancer cells. Hence, targeting lipogenic enzymes and signaling cascades is a very promising approach in developing innovative therapeutic agents for the fight against cancer. This review summarizes main aspects of altered fatty acid synthesis in cancer cells and emphasizes the power of chemical genetic approaches in identifying and analyzing novel anti-cancer drug candidates interfering with lipid metabolism.


Subject(s)
Antineoplastic Agents , Lipogenesis , Neoplasms , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Lipogenesis/physiology , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/physiopathology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
8.
Br J Cancer ; 118(1): 43-51, 2018 01.
Article in English | MEDLINE | ID: mdl-29112683

ABSTRACT

BACKGROUND: Although altered membrane physiology has been discussed within the context of cancer, targeting membrane characteristics by drugs being an attractive therapeutic strategy has received little attention so far. METHODS: Various acetyl-CoA carboxylase 1 (ACC1), and fatty acid synthase (FASN) inhibitors (like Soraphen A and Cerulenin) as well as genetic knockdown approaches were employed to study the effects of disturbed phospholipid composition on membrane properties and its functional impact on cancer progression. By using state-of-the-art methodologies such as LC-MS/MS, optical tweezers measurements of giant plasma membrane vesicles and fluorescence recovery after photobleaching analysis, membrane characteristics were examined. Confocal laser scanning microscopy, proximity ligation assays, immunoblotting as well as migration, invasion and proliferation experiments unravelled the functional relevance of membrane properties in vitro and in vivo. RESULTS: By disturbing the deformability and lateral fluidity of cellular membranes, the dimerisation, localisation and recycling of cancer-relevant transmembrane receptors is compromised. Consequently, impaired activation of growth factor receptor signalling cascades results in abrogated tumour growth and metastasis in different in vitro and in vivo models. CONCLUSIONS: This study highlights the field of membrane properties as a promising druggable cellular target representing an innovative strategy for development of anti-cancer agents.


Subject(s)
Acetyl-CoA Carboxylase/genetics , Enzyme Inhibitors/administration & dosage , Fatty Acid Synthase, Type I/genetics , Lipogenesis/drug effects , Neoplasms/drug therapy , Acetyl-CoA Carboxylase/antagonists & inhibitors , Cell Line, Tumor , Cell Membrane/drug effects , Cell Movement/drug effects , Cell Proliferation , Cerulenin/administration & dosage , Cerulenin/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acid Synthase, Type I/antagonists & inhibitors , Gene Knockdown Techniques , Humans , Macrolides/administration & dosage , Macrolides/pharmacology , Membrane Fluidity/drug effects , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasms/metabolism , Phospholipids/analysis , Photobleaching , Xenograft Model Antitumor Assays
9.
Sci Rep ; 7: 42382, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28186197

ABSTRACT

Marine actinobacteria are drawing more and more attention as a promising source of new natural products. Here we report isolation, genome sequencing and metabolic profiling of new strain Streptomyces sp. MP131-18 isolated from marine sediment sample collected in the Trondheim Fjord, Norway. The 16S rRNA and multilocus phylogenetic analysis showed that MP131-18 belongs to the genus Streptomyces. The genome of MP131-18 isolate was sequenced, and 36 gene clusters involved in the biosynthesis of 18 different types of secondary metabolites were predicted using antiSMASH analysis. The combined genomics-metabolics profiling of the strain led to the identification of several new biologically active compounds. As a result, the family of bisindole pyrroles spiroindimicins was extended with two new members, spiroindimicins E and F. Furthermore, prediction of the biosynthetic pathway for unusual α-pyrone lagunapyrone isolated from MP131-18 resulted in foresight and identification of two new compounds of this family - lagunapyrones D and E. The diversity of identified and predicted compounds from Streptomyces sp. MP131-18 demonstrates that marine-derived actinomycetes are not only a promising source of new natural products, but also represent a valuable pool of genes for combinatorial biosynthesis of secondary metabolites.


Subject(s)
Aquatic Organisms/genetics , Aquatic Organisms/metabolism , Biological Products/analysis , Genomics , Metabolomics , Streptomyces/genetics , Streptomyces/metabolism , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/chemistry , Biosynthetic Pathways , Genome, Bacterial , Indoles/chemistry , Molecular Sequence Annotation , Multigene Family , Phylogeny , Pyrroles/chemistry , Secondary Metabolism/genetics
10.
ChemMedChem ; 12(5): 347-352, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28099785

ABSTRACT

Classic cytotoxic drugs remain indispensable instruments in antitumor therapy due to their effectiveness and a more prevalent insensitivity toward tumor resistance mechanisms. Herein we describe the favorable properties of 6-(N,N-dimethyl-2-aminoethoxy)-11-(3,4,5-trimethoxyphenyl)pyrido[3,4-c][1,9]phenanthroline (P8-D6), a powerful inducer of apoptosis caused by an equipotent inhibition of human topoisomerase I and II activities. A broad-spectrum effect against human tumor cell lines at nanomolar concentrations, as well as strong antileukemic effects, were shown to be superior to those of marketed topoisomerase-targeting drugs and dual topoisomerase inhibitors in clinical trials. The facile four-step synthesis, advantageous drugability properties, and initial in vivo data encourage the application of P8-D6 in appropriate animal tumor models and further drug development.


Subject(s)
Antineoplastic Agents/chemistry , Naphthalenes/chemical synthesis , Topoisomerase I Inhibitors/chemistry , Topoisomerase II Inhibitors/chemistry , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Body Weight/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Nude , Naphthalenes/therapeutic use , Naphthalenes/toxicity , Neoplasms/drug therapy , Topoisomerase I Inhibitors/therapeutic use , Topoisomerase I Inhibitors/toxicity , Topoisomerase II Inhibitors/therapeutic use , Topoisomerase II Inhibitors/toxicity , Transplantation, Heterologous
11.
Adv Healthc Mater ; 6(2)2017 Jan.
Article in English | MEDLINE | ID: mdl-27863166

ABSTRACT

Metal-organic frameworks (MOFs) are promising platforms for the synthesis of nanoparticles for diverse medical applications. Their fundamental design principles allow for significant control of the framework architecture and pore chemistry, enabling directed functionalization for nanomedical applications. However, before applying novel nanomaterials to patients, it is imperative to understand their potential health risks. In this study, the nanosafety of different MOF nanoparticles is analyzed comprehensively for diverse medical applications. The authors first evaluate the effects of MOFs on human endothelial and mouse lung cells, which constitute a first line of defense upon systemic blood-mediated and local lung-specific applications of nanoparticles. Second, we validated these MOFs for multifunctional surface coatings of dental implants using human gingiva fibroblasts. Moreover, biocompatibility of MOFs is assessed for surface coating of nerve guidance tubes using human Schwann cells and rat dorsal root ganglion cultures. The main finding of this study is that the nanosafety and principal suitability of our MOF nanoparticles as novel agents for drug delivery and implant coatings strongly varies with the effector cell type. We conclude that it is therefore necessary to carefully evaluate the nanosafety of MOF nanomaterials with respect to their particular medical application and their interacting primary cell types, respectively.


Subject(s)
Drug Carriers/chemistry , Endothelial Cells/metabolism , Fibroblasts/metabolism , Gingiva/metabolism , Nanoparticles/chemistry , Animals , Drug Carriers/adverse effects , Endothelial Cells/cytology , Fibroblasts/cytology , Gingiva/cytology , Humans , Mice , Nanoparticles/adverse effects
12.
Chem Commun (Camb) ; 51(87): 15752-5, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26359316

ABSTRACT

We report the synthesis of MOF@lipid nanoparticles as a versatile and powerful novel class of nanocarriers based on metal-organic frameworks (MOFs). We show that the MOF@lipid system can effectively store dye molecules inside the porous scaffold of the MOF while the lipid bilayer prevents their premature release. Efficient uptake of the MOF@lipid nanoparticles by cancer cells makes these nanocarriers promising for drug delivery and diagnostic purposes.


Subject(s)
Lipid Bilayers/chemistry , Nanoparticles/chemistry , Organometallic Compounds/chemistry , Cell Line, Tumor , Chromium/chemistry , Drug Carriers , Ferric Compounds/chemistry , Fluorescein/chemistry , Fluorescent Dyes/chemistry , Glycerylphosphorylcholine/analogs & derivatives , Glycerylphosphorylcholine/chemistry , Humans , Phosphatidylcholines , Porosity
13.
Angew Chem Int Ed Engl ; 53(47): 12960-5, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25256790

ABSTRACT

Resistance to chemotherapeutic agents represents a major challenge in cancer research. One approach to this problem is combination therapy, the application of a toxic chemotherapeutic drug together with a sensitizing compound that addresses the vulnerability of cancer cells to induce apoptosis. Here we report the discovery of a new compound class (T8) that sensitizes various cancer cells towards etoposide treatment at subtoxic concentrations. Proteomic analysis revealed protein disulfide isomerase (PDI) as the target of the T8 class. In-depth chemical and biological studies such as the synthesis of optimized compounds, molecular docking analyses, cellular imaging, and apoptosis assays confirmed the unique mode of action through reversible PDI inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Disulfide-Isomerases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Neoplasms/pathology , Protein Disulfide-Isomerases/metabolism , Proteomics , Small Molecule Libraries/chemistry , Structure-Activity Relationship
14.
Biochem Pharmacol ; 91(2): 157-67, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25069048

ABSTRACT

TRAIL (TNFα-related apoptosis-inducing factor) has been promoted as a promising anti-cancer agent. Unfortunately many tumor cells develop resistance towards TRAIL due to numerous defects in apoptotic signaling. To handle this problem combination therapy with compounds affecting as many different anti-apoptotic targets as possible might be a feasible approach. The bromo-substituted indirubin derivative 6BIO meets this challenge: Treatment of breast cancer and bladder carcinoma cell lines with micromolar concentrations of 6BIO abrogates cellular growth and induces apoptosis. Combination of subtoxic amounts of 6BIO with ineffective doses of TRAIL completely abolishes proliferation and long-term survival of cancer cells. As shown in two-dimensional as well as three-dimensional cell culture models, 6BIO potently augments TRAIL-induced apoptosis in cancer cell lines. The potent chemosensitizing effect of 6BIO to TRAIL-mediated cell death is due to the pleiotropic inhibitory profile of 6BIO. As shown previously, 6BIO abrogates STAT3, PDK1 as well as GSK3 signaling and moreover, inhibits the expression of the anti-apoptotic Bcl-2 family members Bcl-xL and Mcl-1 on mRNA as well as on protein level, as demonstrated in this study. Moreover, the expression of cFLIP and cIAP1 is significantly downregulated in 6BIO treated cancer cell lines. In sum (subtoxic concentration of) the multi-kinase inhibitor 6BIO serves as a potent chemosensitizing agent fighting TRAIL resistant cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Oximes/pharmacology , Protein Kinase Inhibitors/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
15.
Bioorg Med Chem ; 22(18): 5117-22, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24679674

ABSTRACT

Natural compounds offer a broad spectrum of potential drug candidates against human malignancies. Several cytostatic drugs, which are in clinical use for decades, derive directly from natural sources or are synthetically optimized derivatives of natural lead structures. An eukaryote target molecule to which many natural derived anti-cancer drugs bind to is the microtubule network. Of similar importance for the cell is the actin cytoskeleton, responsible for cell movements, migration of cells and cytokinesis. Nature provides also a broad range of compounds directed against actin as intracellular target, but none of these actin-targeting compounds has ever been brought to clinical trials. One reason why actin-binding compounds have not yet been considered for further clinical investigations is that little is known about their pharmacological properties in cancer cells. Herein, we focused on the closer characterization of doliculide, an actin binding natural compound of marine origin in the breast cancer cell lines MCF7 and MDA-MB-231. We used fluorescence-recovery-after-photobleaching (FRAP) analysis to determine doliculide's early effects on the actin cytoskeleton and rhodamin-phalloidin staining for long-term effects on the actin CSK. After validating the disruption of the actin network, we further investigated the functional effects of doliculide. Doliculide treatment leads to inhibition of proliferation and impairs the migratory potential. Finally, we could also show that doliculide leads to the induction of apoptosis in both cell lines. Our data for the first time provide a closer characterization of doliculide in breast cancer cells and propagate doliculide for further investigations as lead structure and potential therapeutic option as actin-targeting compound.


Subject(s)
Actins/antagonists & inhibitors , Actins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Depsipeptides/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Binding Sites/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Depsipeptides/chemical synthesis , Depsipeptides/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Molecular Conformation , Protein Binding , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
J Nat Prod ; 77(3): 536-42, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24437936

ABSTRACT

Tubulin binding agents are a potent group of cancer chemotherapeutics. Most of these substances are naturally derived compounds. A novel substance class of destabilizing agents is the group of tubulysins. The tubulysins and their derivative pretubulysin have shown high efficacy in vitro and in vivo. Due to their complex chemical structures, one major bottleneck of the tubulysins is their accessibility. Biotechnological as well as chemical production is challenging, especially on larger scales. Thus, the synthesis of chemically simplified structures is needed with retained or improved biological activity. Herein is presented the biological evaluation of two pretubulysin derivatives [2-desmethylpretubulysin AU816 (1) and phenylpretubulysin JB337 (2)] in comparison to pretubulysin. Both 1 and 2 display a simplification in chemical synthesis. It was shown that both compounds exhibited potent biological activity against cancer cells. These simplified compounds inhibited tubulin polymerization in the nanomolar range. The cytotoxic effects of 1 and 2 were in a similar range, when compared with pretubulysin [IC50 (nM): pretubulysin: 0.6; 1: 10; 2: 100]. Furthermore, it was shown that cell cycle arrest is induced and migration is hampered in MDA-MB-231 breast cancer cells. In conclusion, 1 was shown to be about 10-fold more active than 2 and as potent as pretubulysin.


Subject(s)
Antimitotic Agents/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Angiogenesis Inhibitors/pharmacology , Antimitotic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Humans , Microtubules/drug effects , Microtubules/metabolism , Molecular Structure , Oligopeptides/chemical synthesis , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology
17.
Cancer Res ; 73(19): 6004-12, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23946383

ABSTRACT

While metastasis is the chief cause of cancer mortality, there nonetheless remains a lack of antimetastatic therapies that are clinically available. In this study, we present the indirubin derivative 6-bromo-indirubin-3'-oxime (6BIO) as a promising antimetastatic agent. 6BIO strongly reduced formation of lung metastasis in the well-established 4T1 mouse model of aggressive breast cancer. Several major hallmarks of the metastatic process were affected by subtoxic concentrations of 6BIO, which inhibited adhesion, migration, and invasion of a variety of metastatic cell types in vitro. Mechanistic analyses focused on known targets of 6BIO, which were silenced by this compound. Unexpectedly, RNAi-mediated silencing of glycogen synthase kinase 3ß (GSK3ß) and phosphoinositide-dependent protein kinase 1 (PDK1), both modulators of cellular metastasis targeted by 6BIO, were not found to affect invasive migration in this study. Instead, the Jak/STAT3 signaling pathway appeared to play a major role through modulation of its downstream migration regulators C-terminal tensin-like protein and matrix metalloproteinase 2. However, PDK1 and GSK3ß contributed to the overall response to 6BIO, as silencing of all three pathways resulted in almost complete inhibition of migration, phenocopying the 6BIO response. Taken together, our findings illustrate the antimetastatic activity of 6BIO on the basis of its ability to simultaneously inhibit several kinase cascades involved in metastasis of cancer cells, supporting the concept of "polypharmacology" in developing drugs to attack metastasis, the most deadly aspect of cancer.


Subject(s)
Breast Neoplasms/prevention & control , Cell Movement/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Indoles/pharmacology , Lung Neoplasms/prevention & control , Oximes/pharmacology , Spheroids, Cellular/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Chemotaxis , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , STAT Transcription Factors/metabolism , Signal Transduction
18.
Int J Mol Med ; 28(5): 721-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21743955

ABSTRACT

The repellent factor family of Slit molecules has been described to have repulsive function in the developing nervous system on growing axons expressing the Robo receptors. Alterations of the Slit/Robo system have been observed in various pathological conditions and in cancer. However, until today no detailed studies on Slit function on melanoma migration are available. Therefore, we analysed the mRNA expression in melanoma cells and found induction of Robo3 expression compared to normal melanocytes. Functional assays performed with melanoma cells revealed that treatment with Slit3 led to strong inhibition of migration. Interestingly, we observed down-regulation of AP-1 activity and target gene expression after Slit3 treatment contributing to the negative regulation of migration. Taken together, our data showed that Slit3 reduces the migratory activity of melanoma cells, potentially by repulsion of the cells in analogy to the neuronal system. Further studies will be necessary to prove Slit activity in vivo, but due to its function, Slit3 activity may be helpful in the treatment of melanoma.


Subject(s)
Melanoma/metabolism , Membrane Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Flow Cytometry , Humans , Melanoma/genetics , Mice , Nerve Tissue Proteins/genetics , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Roundabout Proteins
19.
Cell Mol Life Sci ; 67(20): 3535-48, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20480203

ABSTRACT

Since bone morphogenetic proteins (BMPs) play an important role in melanoma progression, we aimed to determine the molecular mechanisms leading to overexpression of BMP4 in melanoma cells compared to normal melanocytes. With our experimental approach we revealed that loss of expression of a microRNA represents the starting point for a signaling cascade finally resulting in overexpression of BMP4 in melanoma cells. In detail, strongly reduced expression of the microRNA miR-196a in melanoma cells compared to healthy melanocytes leads to enhanced HOX-B7 mRNA and protein levels, which subsequently raise Ets-1 activity by inducing basic fibroblast growth factor (bFGF). Ets-1 finally accounts for induction of BMP4 expression. We were furthermore able to demonstrate that bFGF-mediated induction of migration is achieved via activation of BMP4, thus determining BMP4 as major modulator of migration in melanoma. In summary, our study provides insights into the early steps of melanoma progression and might thereby harbor therapeutic relevance.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Melanoma/genetics , MicroRNAs/metabolism , Skin Neoplasms/genetics , Base Sequence , Bone Morphogenetic Protein 4/metabolism , Cell Line, Tumor , Cell Movement , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Homeodomain Proteins/metabolism , Humans , Melanocytes/metabolism , Melanoma/pathology , MicroRNAs/genetics , Models, Biological , Molecular Sequence Data , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Skin Neoplasms/pathology
20.
J Pathol ; 218(4): 520-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19431154

ABSTRACT

Striking similarities exist between molecular mechanisms driving embryonic liver development and progression of hepatocellular carcinoma (HCC). Bone morphogenetic proteins (BMPs), particularly BMP4, have been proposed to regulate embryonic hepatic development. BMP expression has been observed in neoplasia but the expression and biological role of BMP4 in human HCC are unknown. We found increased BMP4 mRNA and protein in HCC cell lines and tissue samples compared to primary human hepatocytes and corresponding non-tumourous tissue. Hypoxia further induced BMP4 expression in HCC cells, which was abolished by transfection of a dominant negative form of HIF-1 alpha (dnHIF-1 alpha). However, gel shift assays revealed only minor binding activity in nuclear extracts from (hypoxic) HCC cells to a putative hypoxia-response element in the BMP4 promoter. Sequence analysis of the BMP4 promoter revealed two Ets-1 binding sites, and Ets-1 activity was increased in HCC cells under hypoxic conditions. Transfection of dnHIF-1 alpha completely abrogated hypoxia-induced Ets-1 activity as well as BMP4 expression. Overexpression of Ets-1 markedly enhanced BMP4 promoter activity, while antisense Ets-1 almost completely abolished basal as well as hypoxia-induced BMP4 expression. These data demonstrate that Ets-1 activity contributes to baseline expression of the BMP4 gene and is the predominant mediator of the HIF-dependent BMP4 induction under hypoxic conditions. To determine the functional relevance of BMP4 expression, HCC cell lines were treated with antisense BMP4 constructs or siRNA against BMP4. BMP4 suppression resulted in a strong reduction of the migratory and invasive potential and anchorage-independent growth. Furthermore, tube formation assays indicated that BMP4 expressed by HCC cells promotes vasculogenesis. Our findings demonstrate that BMP4 is increased in HCC and promotes HCC progression. Therefore, BMP4 expression may have clinical relevance, and interfering with BMP4 signalling appears as an attractive therapeutic target for this highly aggressive tumour.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Bone Morphogenetic Protein 4/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Cell Line, Tumor , Collagen , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Laminin , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic , Proteoglycans , Proto-Oncogene Protein c-ets-1/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...