Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(11): e112542, 2014.
Article in English | MEDLINE | ID: mdl-25391145

ABSTRACT

BACKGROUND: A major player in the process of metastasis is the actin cytoskeleton as it forms key structures in both invasion mechanisms, mesenchymal and amoeboid migration. We tested the actin binding compound Chondramide as potential anti-metastatic agent. METHODS: In vivo, the effect of Chondramide on metastasis was tested employing a 4T1-Luc BALB/c mouse model. In vitro, Chondramide was tested using the highly invasive cancer cell line MDA-MB-231 in Boyden-chamber assays, fluorescent stainings, Western blot and Pull down assays. Finally, the contractility of MDA-MB-231 cells was monitored in 3D environment and analyzed via PIV analysis. RESULTS: In vivo, Chondramide treatment inhibits metastasis to the lung and the migration and invasion of MDA-MB-231 cells is reduced by Chondramide in vitro. On the signaling level, RhoA activity is decreased by Chondramide accompanied by reduced MLC-2 and the stretch induced guanine nucleotide exchange factor Vav2 activation. At same conditions, EGF-receptor autophosphorylation, Akt and Erk as well as Rac1 are not affected. Finally, Chondramide treatment disrupted the actin cytoskeleton and decreased the ability of cells for contraction. CONCLUSIONS: Chondramide inhibits cellular contractility and thus represents a potential inhibitor of tumor cell invasion.


Subject(s)
Antineoplastic Agents/pharmacology , Contractile Proteins/antagonists & inhibitors , Depsipeptides/pharmacology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Peptides, Cyclic/pharmacology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cell Line, Tumor , Cell Movement , Contractile Proteins/genetics , Contractile Proteins/metabolism , Female , Humans , Injections, Intravenous , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
2.
Cardiovasc Res ; 104(2): 303-14, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25239826

ABSTRACT

AIMS: Inhibiting angiogenesis is a major approach in tumour therapy. To combat angiogenesis, the tubulin cytoskeleton has emerged as an interesting target in many pre- and clinical studies. Contrarily, the actin cytoskeleton has been largely neglected as a potential drug target in angiogenesis. However, due to the development of drug resistances, new therapeutic strategies are always needed in tumour treatment. Therefore, the therapeutic potential of actin-binding small molecules is of particular interest. METHODS AND RESULTS: We investigate the impact of chondramide (Ch), an actin polymerizing myxobacterial compound, on angiogenesis and underlying signalling. Chondramide treatment not only reduces the migration of endothelial cells but also the maturation of endothelial tube networks on matrigel. These observations can partly be explained by a disintegration of stress fibres due to aggregation and subsequent accumulation of actin in cellular structures known as 'aggresomes'. Chondramide treatment impairs the maturation of focal adhesions and reduces the amount of active ß1 integrin at the cell surface. Accordingly, signalling events downstream of focal adhesions are reduced. Thus, we observed that the activity of Src and downstream factors Rho-GTPases Rac1 and Rho is reduced upon Ch treatment. In vivo, Ch was well tolerated in mice and vascularization of a tumour xenograft as well as of the developing retina was significantly reduced. CONCLUSION: Chondramide diminishes angiogenesis via two ways: (i) the disintegration of stress fibres and (ii) the reduction of promigratory signals. Our findings highlight Ch as a novel class of therapeutic lead compound with anti-angiogenic potential.


Subject(s)
Actin Cytoskeleton/drug effects , Angiogenesis Inhibitors/pharmacology , Bacterial Proteins/pharmacology , Breast Neoplasms/drug therapy , Depsipeptides/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Pathologic , Neovascularization, Physiologic/drug effects , Actin Cytoskeleton/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Integrin beta1/metabolism , Mice, SCID , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , Time Factors , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
3.
Mol Cancer Ther ; 13(4): 926-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24482380

ABSTRACT

Fighting metastasis is a major challenge in cancer therapy and novel therapeutic targets and drugs are highly appreciated. Resistance of invasive cells to anoikis, a particular type of apoptosis induced by loss of cell-matrix contact, is a major prerequisite for their metastatic spread. Inducing anoikis in metastatic cancer cells is therefore a promising therapeutic approach. The vacuolar-ATPase (V-ATPase), a proton pump located at the membrane of acidic organelles, has recently come to focus as an antimetastatic cancer target. As V-ATPase inhibitors have shown to prevent invasion of tumor cells and are able to induce apoptosis, we proposed that V-ATPase inhibition induces anoikis-related pathways in invasive cancer cells. We used the V-ATPase inhibitor archazolid to investigate the mechanism of anoikis induction in various metastatic cancer cells (T24, MDA-MB-231, 4T1, 5637) in vitro. Anoikis induction by archazolid was characterized by decreased c-FLIP expression and caspase-8 activation as well as reduction of active integrin-ß1 and an early increase of the proapoptotic protein BIM. However, we observed that archazolid also induces mechanisms opposing anoikis such as degradation of BIM mediated by extracellular signal-regulated kinase (ERK), Akt and Src kinases at later time points and induction of reactive oxygen species. Still, intravenous injection of archazolid-treated 4T1-Luc2 mouse breast cancer cells resulted in reduced metastasis in mouse lungs. Thus, V-ATPase inhibition is not only an interesting option to reduce cancer metastasis, but also to better understand anoikis resistance and to find choices to fight against it.


Subject(s)
Anoikis/drug effects , Breast Neoplasms/drug therapy , Liver Neoplasms, Experimental/drug therapy , Macrolides/pharmacology , Neoplasm Invasiveness/genetics , Thiazoles/pharmacology , Urinary Bladder Neoplasms/drug therapy , Animals , Anoikis/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms, Experimental/secondary , Macrolides/therapeutic use , Male , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Thiazoles/therapeutic use , Urinary Bladder Neoplasms/pathology
4.
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
5.
Int J Cancer ; 134(10): 2478-88, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24166050

ABSTRACT

The myxobacterial agent archazolid inhibits the vacuolar proton pump V-ATPase. V-ATPases are ubiquitously expressed ATP-dependent proton pumps, which are known to regulate the pH in endomembrane systems and thus play a crucial role in endo- and exocytotic processes of the cell. As cancer cells depend on a highly active secretion of proteolytic proteins in order to invade tissue and form metastases, inhibition of V-ATPase is proposed to affect the secretion profile of cancer cells and thus potentially abrogate their metastatic properties. Archazolid is a novel V-ATPase inhibitor. Here, we show that the secretion pattern of archazolid treated cancer cells includes various prometastatic lysosomal proteins like cathepsin A, B, C, D and Z. In particular, archazolid induced the secretion of the proforms of cathepsin B and D. Archazolid treatment abrogates the cathepsin B maturation process leading to reduced intracellular mature cathepsin B protein abundance and finally decreased cathepsin B activity, by inhibiting mannose-6-phoshate receptor-dependent trafficking. Importantly, in vivo reduced cathepsin B protein as well as a decreased proteolytic cathepsin B activity was detected in tumor tissue of archazolid-treated mice. Our results show that inhibition of V-ATPase by archazolid reduces the activity of prometastatic proteases like cathepsin B in vitro and in vivo.


Subject(s)
Cathepsin B/metabolism , Macrolides/pharmacology , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Blotting, Western , Cathepsin B/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/drug effects , Lysosomes/enzymology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/prevention & control , Protein Transport/drug effects , RNA Interference , Receptor, IGF Type 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
6.
Transl Oncol ; 6(1): 1-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23418611

ABSTRACT

Resistance formation is one of the major hurdles in cancer therapy. Metronomic anti-angiogenic treatment of xenografted prostate cancer tumors in severe combined-immunodeficiency (SCID) mice with cyclophosphamide (CPA) results in the appearance of resistant tumors. To investigate the complex molecular changes occurring during resistance formation, we performed a comprehensive gene expression analysis of the resistant tumors in vivo. We observed a multitude of differentially expressed genes, e.g., PAS domain containing protein 1, annexin A3 (ANXA3), neurotensin, or plasminogen activator tissue (PLAT), when comparing resistant to in vivo passaged tumor samples. Furthermore, tumor cells from in vivo and in vitro conditions showed a significant difference in target gene expression. We assigned the differentially expressed genes to functional pathways like axon guidance, steroid biosynthesis, and complement and coagulation cascades. Most of these genes were involved in anti-coagulation. Up-regulation of anticoagulatory ANXA3 and PLAT and down-regulation of PLAT inhibitor serpin peptidase inhibitor clade A were validated by quantitative real-time polymerase chain reaction. In contrast, coagulation factor F3 was upregulated, accompanied by the expression of an altered gene product. These findings give insights into the resistance mechanisms of metronomic CPA treatment, suggesting an important role of anti-coagulation in resistance formation.

7.
Cancer Res ; 72(22): 5976-87, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22986742

ABSTRACT

The abundance of the multimeric vacuolar ATP-dependent proton pump, V-ATPase, on the plasma membrane of tumor cells correlates with the invasiveness of the tumor cell, suggesting the involvement of V-ATPase in tumor metastasis. V-ATPase is hypothesized to create a proton efflux leading to an acidic pericellular microenvironment that promotes the activity of proinvasive proteases. An alternative, not yet explored possibility is that V-ATPase regulates the signaling machinery responsible for tumor cell migration. Here, we show that pharmacologic or genetic reduction of V-ATPase activity significantly reduces migration of invasive tumor cells in vitro. Importantly, the V-ATPase inhibitor archazolid abrogates tumor dissemination in a syngeneic mouse 4T1 breast tumor metastasis model. Pretreatment of cancer cells with archazolid impairs directional motility by preventing spatially restricted, leading edge localization of epidermal growth factor receptor (EGFR) as well as of phosphorylated Akt. Archazolid treatment or silencing of V-ATPase inhibited Rac1 activation, as well as Rac1-dependent dorsal and peripheral ruffles by inhibiting Rab5-mediated endocytotic/exocytotic trafficking of Rac1. The results indicate that archazolid effectively decreases metastatic dissemination of breast tumors by impairing the trafficking and spatially restricted activation of EGFR and Rho-GTPase Rac1, which are pivotal for directed movement of cells. Thus, our data reveals a novel mechanism underlying the role of V-ATPase in tumor dissemination.


Subject(s)
Breast Neoplasms/drug therapy , Macrolides/pharmacology , Thiazoles/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Polarity/drug effects , Down-Regulation , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Xenograft Model Antitumor Assays , rab5 GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism
8.
Sensors (Basel) ; 12(3): 3370-93, 2012.
Article in English | MEDLINE | ID: mdl-22737014

ABSTRACT

Pollution of drinking water sources represents a continuously emerging problem in global environmental protection. Novel techniques for real-time monitoring of water quality, capable of the detection of unanticipated toxic and bioactive substances, are urgently needed. In this study, the applicability of a cell-based sensor system using selected eukaryotic cell lines for the detection of aquatic pollutants is shown. Readout parameters of the cells were the acidification (metabolism), oxygen consumption (respiration) and impedance (morphology) of the cells. A variety of potential cytotoxic classes of substances (heavy metals, pharmaceuticals, neurotoxins, waste water) was tested with monolayers of L6 cells (rat myoblasts). The cytotoxicity or cellular effects induced by inorganic ions (Ni(2+) and Cu(2+)) can be detected with the metabolic parameters acidification and respiration down to 0.5 mg/L, whereas the detection limit for other substances like nicotine and acetaminophen are rather high, in the range of 0.1 mg/L and 100 mg/L. In a close to application model a real waste water sample shows detectable signals, indicating the existence of cytotoxic substances. The results support the paradigm change from single substance detection to the monitoring of overall toxicity.


Subject(s)
Drinking Water/chemistry , Environmental Monitoring , Water Pollutants, Chemical/toxicity , Acetaminophen/chemistry , Acetaminophen/toxicity , Animals , Cell Line , Copper Sulfate/chemistry , Copper Sulfate/toxicity , Cricetinae , Electric Impedance , HT29 Cells , Hep G2 Cells , Humans , Nickel/chemistry , Nickel/toxicity , Nicotine/chemistry , Nicotine/toxicity , Rats , Water Pollutants, Chemical/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...