Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Sci Rep ; 8(1): 18042, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575776

ABSTRACT

Cells vary in their dynamic response to external stimuli, due to stochastic fluctuations and non-uniform progression through the cell cycle. Hence, single-cell studies are required to reveal the range of heterogeneity in their responses to defined perturbations, which provides detailed insight into signaling processes. Here, we present a time-lapse study using arrays of micro-trenches to monitor the timing of cell division and apoptosis in non-adherent cells at the single-cell level. By employing automated cell tracking and division detection, we precisely determine cell cycle duration and sister-cell correlations for hundreds of individual cells in parallel. As a model application we study the response of leukemia cells to the chemostatic drug vincristine as a function of cell cycle phase. The time-to-death after drug addition is found to depend both on drug concentration and cell cycle phase. The resulting timing and dose-response distributions were reproduced in control experiments using synchronized cell populations. Interestingly, in non-synchronized cells, the time-to-death intervals for sister cells appear to be correlated. Our study demonstrates the practical benefits of micro-trench arrays as a platform for high-throughput, single-cell time-lapse studies on cell cycle dependence, correlations and cell fate decisions in general.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Culture Techniques , Cell Division/drug effects , Drug Screening Assays, Antitumor , Single-Cell Analysis , Antineoplastic Agents/isolation & purification , Automation, Laboratory , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Tracking , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Humans , Microtechnology/instrumentation , Microtechnology/methods , Single-Cell Analysis/instrumentation , Single-Cell Analysis/methods , Time-Lapse Imaging/instrumentation , Time-Lapse Imaging/methods , Tissue Scaffolds/chemistry , Tumor Cells, Cultured
2.
Br J Pharmacol ; 173(17): 2645-56, 2016 09.
Article in English | MEDLINE | ID: mdl-27390037

ABSTRACT

BACKGROUND AND PURPOSE: Cyclin-dependent kinase 5 (CDK5) has recently emerged as an attractive target in several tumour entities. Inhibition of CDK5 has been shown to have anti-angiogenic effects in vitro and in vivo. However, potent inhibitors of CDK5, which can be applied in vivo, are still scarce. We have recently developed a new series of 5-substituted 3-isopropyl-7-[4-(2-pyridyl)benzyl]amino-1(2)H-pyrazolo[4,3-d]pyrimidines that show a preference for inhibiting CDK5 and tested them in vitro and in vivo in a murine model of hepatocellular carcinoma. EXPERIMENTAL APPROACH: All compounds were initially examined for effects on proliferation of HUVECs. The most potent compounds were then tested on migration, and one of them, LGR2674, was selected for assessing effects on nuclear fragmentation, cell cycle, cell viability and metabolic activity. Furthermore, LGR2674 was tested in a tube formation assay and in vivo in a murine model of hepatocellular carcinoma, induced by s.c. injection of HUH7 cells (measurement of in vivo toxicity, tumour vascularization, tumour cell proliferation and tumour size). KEY RESULTS: LGR2674 showed an EC50 in the low nanomolar range in the proliferation and migration assays. Cytotoxic effects started at 50 nM, a concentration that did not influence the cell cycle. In vivo, LGR2674 was well tolerated and caused a clear reduction in vessel density in the tumours; also tumour cell proliferation was inhibited and tumour growth retarded. CONCLUSIONS AND IMPLICATIONS: Pyrazolo[4,3-d]pyrimidine is a novel scaffold for the development of potent CDK inhibitors with in vivo potential. Such structures are good candidates for broadening our pharmacological arsenal against various tumours.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Angiogenesis Inhibitors/chemical synthesis , Animals , Antineoplastic Agents/chemical synthesis , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Mice , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
3.
Pharmazie ; 70(9): 610-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26492647

ABSTRACT

Several natural compound interfere with microtubules or the actin cytoskeleton. Compounds interfering with the microtubules like Vinca-alkaloids or taxanes, are extensively used for cancer therapy. In contrast, knowledge about pharmacological properties of actin binding drugs is poor and drugs interfering with actin are far from clinical use. Rhizopodin is a natural compound that strongly affects the actin cytoskeleton at nanomolar concentrations. Initial work revealed interesting anti-bacterial and cytotoxic effects, but the cellular effects and pharmacological properties of rhizopodin have not been characterized. We hypothesized that rhizopodin might exert anti-cancer activity. Therefore, the aim of this study was to characterize the cellular and pharmacological effects of rhizopodin in cancer. Effects of rhizopodin demonstrated prominent effects on the actin cytoskeleton as shown in the actin-pyrene assay and by immunostaining of cancer cells. To investigate cellular effects of rhizopodin, we analyzed cell proliferation, cell death induction by propidium iodide exclusion and western blot, as well as migration by impedance measurement using the xCELLligence device in MDA-MB-231 breast cancer and T24 bladder cancer cell lines. Rhizopodin inhibited proliferation and induced cell death of MDA-MB-231 and T24 cells at nanomolar concentrations. PARP cleavage by rhizopodin suggests caspase-dependent cell death induction. Importantly, rhizopodin potently inhibited MDA-MB-231 and T24 cancer cell migration at subtoxic doses where no actin aggregation was observed, indicating a specific underlying signaling of rhizopodin. In summary, our study elucidates rhizopodin as actin-binding natural compound that exerts potent anti-cancer effects. Therefore, our work provides the basis for further in depth characterization of rhizopodin as an antitumoral agent.


Subject(s)
Actins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Macrolides/pharmacology , Oxazoles/pharmacology , Cell Death/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cytoskeleton/drug effects , Female , Humans
4.
Cell Death Dis ; 5: e1398, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25165884

ABSTRACT

Targeting the actin cytoskeleton (CSK) of cancer cells offers a valuable strategy in cancer therapy. There are a number of natural compounds that interfere with the actin CSK, but the mode of their cytotoxic action and, moreover, their tumor-specific mechanisms are quite elusive. We used the myxobacterial compound Chondramide as a tool to first elucidate the mechanisms of cytotoxicity of actin targeting in breast cancer cells (MCF7, MDA-MB-231). Chondramide inhibits cellular actin filament dynamics shown by a fluorescence-based analysis (fluorescence recovery after photobleaching (FRAP)) and leads to apoptosis characterized by phosphatidylserine exposure, release of cytochrome C from mitochondria and finally activation of caspases. Chondramide enhances the occurrence of mitochondrial permeability transition (MPT) by affecting known MPT modulators: Hexokinase II bound to the voltage-dependent anion channel (VDAC) translocated from the outer mitochondrial membrane to the cytosol and the proapoptotic protein Bad were recruited to the mitochondria. Importantly, protein kinase C-ɛ (PKCɛ), a prosurvival kinase possessing an actin-binding site and known to regulate the hexokinase/VDAC interaction as well as Bad phosphorylation was identified as the link between actin CSK and apoptosis induction. PKCɛ, which was found overexpressed in breast cancer cells, accumulated in actin bundles induced by Chondramide and lost its activity. Our second goal was to characterize the potential tumor-specific action of actin-binding agents. As the nontumor breast epithelial cell line MCF-10A in fact shows resistance to Chondramide-induced apoptosis and notably express low level of PKCɛ, we suggest that trapping PKCɛ via Chondramide-induced actin hyperpolymerization displays tumor cell specificity. Our work provides a link between targeting the ubiquitously occurring actin CSK and selective inhibition of pro-tumorigenic PKCɛ, thus setting the stage for actin-stabilizing agents as innovative cancer drugs. This is moreover supported by the in vivo efficacy of Chondramide triggered by abrogation of PKCɛ signaling shown in a xenograft breast cancer model.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Protein Kinase C/metabolism , Actin Cytoskeleton/drug effects , Actins/genetics , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , Depsipeptides/therapeutic use , Depsipeptides/toxicity , Female , Fluorescence Recovery After Photobleaching , Hexokinase/metabolism , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , Mitochondria/metabolism , Phosphatidylserines/pharmacology , Signal Transduction , Transplantation, Heterologous , Voltage-Dependent Anion Channels/metabolism , bcl-Associated Death Protein/metabolism
5.
Cell Death Dis ; 5: e1001, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24434509

ABSTRACT

Tubulin-binding agents such as taxol, vincristine or vinblastine are well-established drugs in clinical treatment of metastatic cancer. However, because of their highly complex chemical structures, the synthesis and hence the supply issues are still quite challenging. Here we set on stage pretubulysin, a chemically accessible precursor of tubulysin that was identified as a potent microtubule-binding agent produced by myxobacteria. Although much simpler in chemical structure, pretubulysin abrogates proliferation and long-term survival as well as anchorage-independent growth, and also induces anoikis and apoptosis in invasive tumor cells equally potent to tubulysin. Moreover, pretubulysin posseses in vivo efficacy shown in a chicken chorioallantoic membrane (CAM) model with T24 bladder tumor cells, in a mouse xenograft model using MDA-MB-231 mammary cancer cells and finally in a model of lung metastasis induced by 4T1 mouse breast cancer cells. Pretubulysin induces cell death via the intrinsic apoptosis pathway by abrogating the expression of pivotal antiapoptotic proteins, namely Mcl-1 and Bcl-xL, and shows distinct chemosensitizing properties in combination with TRAIL in two- and three-dimensional cell culture models. Unraveling the underlying signaling pathways provides novel information: pretubulysin induces proteasomal degradation of Mcl-1 by activation of mitogen-activated protein kinase (especially JNK (c-Jun N-terminal kinase)) and phosphorylation of Mcl-1, which is then targeted by the SCF(Fbw7) E3 ubiquitin ligase complex for ubiquitination and degradation. In sum, we designate the microtubule-destabilizing compound pretubulysin as a highly promising novel agent for mono treatment and combinatory treatment of invasive cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Female , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/physiopathology
6.
Pharmazie ; 68(7): 478-83, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23923626

ABSTRACT

Microtubule-targeting agents (MTAs) are the most frequently used anti-cancer drugs. They can be divided into tubulin stabilizing and destabilizing agents. Their mode of action has been ascribed to their ability to interfere with the spindle apparatus and, thus, to block mitosis leading to tumor cell death. However, this view has been challenged in the last years and it became increasingly evident that non-mitotic actions of MTAs, i.e. their ability to affect the dynamics of interphase microtubules, are the most relevant mechanism underlying their efficacy. In this review we are presenting a distinct selection of examples of studies describing biological effects of MTAs in three areas: (i) mitosis-independent cell death and metastasis, (ii) tumor angiogenesis, and (iii) vascular-disrupting activity.


Subject(s)
Antineoplastic Agents , Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Cell Death/drug effects , Humans , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Neoplasm Metastasis/prevention & control , Neoplasms/blood supply , Regional Blood Flow/drug effects , Tubulin/metabolism
7.
Br J Pharmacol ; 167(5): 1048-61, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22595030

ABSTRACT

BACKGROUND AND PURPOSE: The use of tubulin-binding compounds, which act in part by inhibiting tumour angiogenesis, has become an integral strategy of tumour therapy. Recently, tubulysins were identified as a novel class of natural compounds of myxobacterial origin, which inhibit tubulin polymerization. As these compounds are structurally highly complex, the search for simplified precursors [e.g. pretubulysin (Prt)] and their derivatives is mandatory to overcome supply problems hampering clinical development. We tested the anti-angiogenic efficacy of Prt and seven of its derivatives in comparison to tubulysin A (TubA). EXPERIMENTAL APPROACH: The compounds were tested in cellular angiogenesis assays (proliferation, cytotoxicity, cell cycle, migration, chemotaxis, tube formation) and in vitro (tubulin polymerization). The efficacy of Prt was also tested in vivo in a murine subcutaneous tumour model induced with HUH7 cells; tumour size and vascularization were measured. KEY RESULTS: The anti-angiogenic potency of all the compounds tested ran parallel to their inhibition of tubulin polymerization in vitro. Prt showed nearly the same efficacy as TubA (EC(50) in low nanomolar range in all cellular assays). Some modifications in the Prt molecule caused only a moderate drop in potency, while others resulted in a dramatic loss of action, providing initial insight into structure-activity relations. In vivo, Prt completely prevented tumour growth and reduced vascular density to 30%. CONCLUSIONS AND IMPLICATIONS: Prt, a chemically accessible precursor of some tubulysins is a highly attractive anti-angiogenic compound both in vitro and in vivo. Even more simplified derivatives of this compound still retain high anti-angiogenic efficacy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Oligopeptides/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotaxis/drug effects , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, SCID , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Oligopeptides/therapeutic use , Tubulin/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Gut ; 58(12): 1670-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19470497

ABSTRACT

BACKGROUND AND AIMS: The transcription factor nuclear factor kappa B (NF-kappaB) has risen as a promising target for anti-inflammatory therapeutics. In the liver, however, NF-kappaB inhibition mediates both damaging and protective effects. The outcome is deemed to depend on the liver cell type addressed. Recent gene knock-out studies focused on the role of NF-kappaB in hepatocytes, whereas the role of NF-kappaB in Kupffer cells has not yet been investigated in vivo. Here we present a novel approach, which may be suitable for clinical application, to selectively target NF-kappaB in Kupffer cells and analyse the effects in experimental models of liver injury. METHODS: NF-kappaB inhibiting decoy oligodeoxynucleotides were loaded upon gelatin nanoparticles (D-NPs) and their in vivo distribution was determined by confocal microscopy. Liver damage, NF-kappaB activity, cytokine levels and apoptotic protein expression were evaluated after lipopolysaccharide (LPS), d-galactosamine (GalN)/LPS, or concanavalin A (ConA) challenge and partial warm ischaemia and subsequent reperfusion, respectively. RESULTS: D-NPs were selectively taken up by Kupffer cells and inhibited NF-kappaB activation. Inhibition of NF-kappaB in Kupffer cells improved survival and reduced liver injury after GalN/LPS as well as after ConA challenge. While anti-apoptotic protein expression in liver tissue was not reduced, pro-apoptotic players such as cJun N-terminal kinase (JNK) were inhibited. In contrast, selective inhibition of NF-kappaB augmented reperfusion injury. CONCLUSIONS: NF-kappaB inhibiting decoy oligodeoxynucleotide-loaded gelatin nanoparticles is a novel tool to selectively inhibit NF-kappaB activation in Kupffer cells in vivo. Thus, liver injury can be reduced in experimental fulminant hepatitis, but increased at ischaemia-reperfusion.


Subject(s)
Kupffer Cells/metabolism , Liver Failure, Acute/prevention & control , NF-kappa B/antagonists & inhibitors , Oligodeoxyribonucleotides/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis , Disease Models, Animal , Gelatin/metabolism , Genetic Therapy/methods , Liver/blood supply , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Male , Mice , NF-kappa B/metabolism , Nanoparticles , Oligodeoxyribonucleotides/pharmacokinetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Translocation, Genetic
9.
Cell Death Differ ; 15(12): 1930-40, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18806754

ABSTRACT

Cephalostatin 1 is a natural compound isolated from a marine worm that induces apoptosis in tumor cells via an apoptosome-independent but caspase-9-dependent pathway and through an endoplasmic reticulum stress response that is accompanied by caspase-4 activation. Here, we show that cephalostatin evokes mitochondrial Smac (second mitochondria-derived activator of caspases) but not cytochrome c release in various carcinoma cell lines. We also show that Smac is critically involved in caspase-9 activation as evidenced by gene silencing experiments. Remarkably, caspase-2 appears to be a major target for cephalostatin-induced cytosolic Smac. Using biochemical and genetic inhibition experiments, we demonstrate that caspase-2 participates in the apoptotic machinery induced by cephalostatin. Cephalostatin-activated caspase-2 appears to act as initiator caspase and is not involved in the activation of caspase-9. Importantly, experiments immunoprecipitating PIDD (p53-induced protein with a DD), RAIDD (RIP-associated ICH-1/CED-3-homologous protein with DD) and caspase-2 identify cephalostatin as an experimental drug that induces the formation of the PIDDosome. The bis-steroid cephalostatin proves to be both a helpful tool to investigate apoptotic signaling and a promising chemotherapeutic agent.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Phenazines/pharmacology , Spiro Compounds/pharmacology , Steroids/pharmacology , Apoptosis Regulatory Proteins , Calpain/metabolism , Carrier Proteins/metabolism , Caspase 2/metabolism , Caspase 9/metabolism , Cell Death/drug effects , Cytochromes c/metabolism , Death Domain Receptor Signaling Adaptor Proteins , Enzyme Activation/drug effects , Gene Silencing/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Jurkat Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects
10.
Leukemia ; 22(9): 1737-45, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18548102

ABSTRACT

Spongistatin 1 is a new experimental chemotherapeutic agent isolated from marine sponges. Here we show that spongistatin 1 potently induces cell death in patient primary acute leukemic cells with higher efficiency than 8/10 clinically used cytotoxic drugs and prevents long-term survival of leukemic cell lines. Spongistatin 1 triggers caspase-dependent apoptosis in Jurkat T cells by the release of cytochrome c, Smac/DIABLO and Omi/HtrA2. As caspase-9 acts as an initiator caspase and Bcl-2 and Bcl-xL overexpression suppress spongistatin 1-induced apoptosis, cell death is mediated through the mitochondrial apoptosis pathway. Importantly, spongistatin 1 leads to the degradation of the antiapoptotic X-linked inhibitor of apoptosis protein. In apoptosis-resistant leukemic tumor cells overexpressing XIAP, spongistatin 1 effectively causes cell death and potentiates cell death induction by other apoptosis-promoting factors that might be caused by spongistatin 1-mediated degradation of XIAP. Our data show that spongistatin 1 represents a promising novel therapeutic agent for the treatment of leukemic tumor cells especially in the clinically highly relevant situation of chemoresistance due to overexpression of XIAP.


Subject(s)
Leukemia/drug therapy , Macrolides/pharmacology , X-Linked Inhibitor of Apoptosis Protein/drug effects , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Humans , Leukemia/pathology , Macrolides/therapeutic use , Mitochondrial Proteins , Porifera , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein/metabolism
11.
Cell Mol Life Sci ; 64(13): 1715-22, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17497242

ABSTRACT

Beneficial effects of Ginkgo biloba on peripheral arterial occlusive disease have been repeatedly shown in clinical trials, especially after use of EGb 761, a standardized special extract. Since the underlying mechanisms are widely unknown, we aimed to elucidate the molecular basis on which EGb 761 protects against endothelial dysfunction in vitro and in vivo. Application of therapeutically feasible doses of EGb 761 for 48 h caused endothelial nitric oxide (NO) production by increasing endothelial nitric oxide synthase (eNOS) promoter activity and eNOS expression in vitro. Phosphorylation of eNOS at a site typical for Akt (Ser 1177) was acutely enhanced by treatment with EGb 761, as was Akt phosphorylation at Ser 478. Furthermore, the extract caused acute relaxation of isolated aortic rings and NO-dependent reduction of blood pressure in vivo in rats. These influences on eNOS represent a putative molecular basis for the protective cardiovascular properties of EGb 761.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nitric Oxide/biosynthesis , Plant Extracts/pharmacology , Animals , Aorta, Thoracic/enzymology , Blood Pressure/drug effects , Cell Line , Endothelial Cells/cytology , Enzyme Activation/drug effects , Ginkgo biloba , Humans , In Vitro Techniques , Male , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Systole/drug effects , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics , Vasodilation/drug effects
12.
Leukemia ; 16(1): 74-83, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11840266

ABSTRACT

The organosulfur compound ajoene, a constitutent of garlic, has been shown to induce apoptosis in a leukemic cell line as well as in blood cells of a leukemic patient. The mechanisms of action of ajoene, however, are unknown. The present study aims to characterize the molecular events leading to ajoene-triggered apoptosis. We show here that ajoene (20 microM) leads to a time-dependent activation of caspase-3-like activity as well as to the proteolytic processing of procaspase-3 and -8. Activation of caspases was necessary for ajoene-induced apoptosis since the broad-range caspase inhibitor zVAD-fmk completely abrogated ajoene-mediated DNA fragmentation. Although the initiator caspase-8 was activated, the CD95 death receptor was not involved in death signaling since the HL-60 clone used was shown to express a functionally inactive CD95 receptor. Furthermore, ajoene induced the release of cytochrome c, which was not inhibited by zVAD-fmk indicating that cytochrome c release precedes caspase activation. Ajoene also led to a dissipation of the mitochondrial transmembrane potential. Overexpression of Bcl-x(L) clearly diminished ajoene-induced caspase activation as well as apoptosis. These results indicate that apoptosis in leukemia cells triggered by ajoene is based on the activation of a mitochondria-dependent caspase cascade which includes also the activation of the initiator caspase-8.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Disulfides/pharmacology , HL-60 Cells/drug effects , Mitochondria/drug effects , Plant Extracts/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/analysis , DNA Fragmentation/drug effects , DNA, Neoplasm/drug effects , Disulfides/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Precursors/metabolism , Fas Ligand Protein , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells/metabolism , Humans , Intracellular Membranes/drug effects , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Membrane Glycoproteins/physiology , Membrane Potentials/drug effects , Mitochondria/enzymology , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidative Stress , Permeability/drug effects , Plant Extracts/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species , Sulfoxides , bcl-X Protein , fas Receptor/biosynthesis , fas Receptor/genetics
13.
FEBS Lett ; 506(2): 131-4, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11591386

ABSTRACT

4,5-Diaminofluorescein (DAF-2) and its membrane-permeable derivate DAF-2 diacetate are fluorescent probes that have been developed to perform real-time biological detection of nitric oxide (NO). Their use for intracellular imaging, however, has recently been seriously questioned and data using DAF-2 for extracellular NO detection at low levels, as for example released from endothelial cells, are rare. Here we show that a reliable detection of low levels of NO in biological systems by DAF-2 is possible (a) by using low DAF-2 concentrations (0.1 microM) and (b) by subtracting the DAF-2 auto-fluorescence from the measured total fluorescence. The described method allows easy real-time detection of endothelial NO formation.


Subject(s)
Endothelium, Vascular/chemistry , Fluorescein , Nitric Oxide/analysis , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Humans , Molecular Structure , Spectrometry, Fluorescence , Tetradecanoylphorbol Acetate/pharmacology , Triazenes/metabolism , omega-N-Methylarginine/pharmacology
14.
Mol Pharmacol ; 60(3): 421-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502871

ABSTRACT

BACKGROUND: The inducible nitric-oxide synthase (iNOS) synthesizes NO from L-arginine. Availability of L-arginine is maintained by a lipopolysaccharide (LPS)-induced induction of the CAT-2B amino acids transporter. Recently, we could show that the cardiovascular hormone atrial natriuretic peptide (ANP) inhibits the induction of iNOS in LPS-stimulated macrophages via its guanylate cyclase-coupled A-receptor. PURPOSE: To investigate whether ANP exerts an effect on LPS-induced L-arginine uptake. METHODS: Murine bone marrow derived macrophages were activated with LPS (1 microg/ml, 20 h) in the presence or absence of ANP or C-type natriuretic peptide (CNP). L-Arginine transport was determined by measuring the uptake of L-[(3)H]arginine. L-[(3)H]Arginine influx was also determined in LPS-activated cells in the presence of N(G)-monomethyl-L-arginine (L-NMMA), competitor amino acids, or ANP. Nitrite accumulation was determined in supernatants of LPS-activated cells cultured in the presence or absence of L-ornithine. RESULTS: ANP dose dependently (10(-8)-10(-6)M) inhibited LPS-induced L-[(3)H]arginine uptake when added simultaneously with LPS, whereas it showed no effect when added simultaneously with L-[(3)H]arginine. The effect was abrogated by the A-receptor antagonist HS-142-1 (10 microg/ml). CNP (10(-6) M) did not influence L-arginine transport. Competitor amino acids (10(-2) M) inhibited L-[(3)H]arginine uptake. An excess of unlabeled L-arginine (10(-2) M) as well as its analog L-NMMA (10(-3) M) also reduced L-[(3)H]arginine influx. L-Arginine uptake was critical for production of NO because L-ornithine significantly decreased LPS-induced nitrite accumulation. CONCLUSION: This work demonstrates that ANP inhibits LPS-induced L-arginine uptake via its guanylate cyclase-coupled A-receptor. Besides its influence on the induction of iNOS, this effect may represent an important and unique mechanism by which ANP regulates NO production in macrophages.


Subject(s)
Arginine/metabolism , Atrial Natriuretic Factor/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Binding, Competitive , Biological Transport/drug effects , Cells, Cultured , Drug Interactions , Guanylate Cyclase/metabolism , Hormones/pharmacology , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide Synthase Type II , Receptors, Atrial Natriuretic Factor/metabolism , Substrate Specificity , Tritium
15.
Hepatology ; 34(3): 535-47, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526540

ABSTRACT

Liver resident NK1.1+ T cells are supposed to play a pivotal role in the onset of inflammatory liver injury in experimental mouse models such as concanavalin A (Con A)-induced hepatitis. These cells, expressing the adhesion receptor, CD44, are largely depleted from the liver by a single intravenous injection of low-molecular-weight fragments of hyaluronic acid (LMW-HA). Here, we report that LMW-HA pretreatment protected mice from liver injury in several models of T-cell- and macrophage-dependent, tumor necrosis factor alpha (TNF-alpha)-mediated inflammatory liver injury, i.e., from liver injury induced by either Con A or Pseudomonas exotoxin A (PEA) or PEA/lipopolysaccharide (LPS). Interestingly, apart from inhibition of cellular adhesion, pretreatment of mice with LMW-HA was also capable of preventing hepatocellular apoptosis and activation of caspase-3 induced by direct administration of recombinant murine (rmu) TNF-alpha to D-galactosamine (GalN)-sensitized mice. LMW-HA-induced hepatoprotection could be neutralized by pretreatment with the nuclear factor-kappaB (NF-kappaB) inhibitor, pyrrolidine dithiocarbamate (PDTC), demonstrating the involvement of NF-kappaB in the observed protective mechanism. Indeed, injection of LMW-HA rapidly induced the production of TNF-alpha by Kupffer cells and the translocation of NF-kappaB into hepatocellular nuclei. Both LMW-HA-induced TNF-alpha production and NF-kappaB translocation were blocked by pretreatment with PDTC. Our findings provide evidence for an unknown mechanism of LMW-HA-dependent protection from inflammatory liver disease, i.e., induction of TNF-alpha- and NF-kappaB-dependent cytoprotective proteins within the target parenchymal liver cells.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Chemical and Drug Induced Liver Injury/prevention & control , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , NF-kappa B/physiology , Tumor Necrosis Factor-alpha , Virulence Factors , Animals , CD4 Lymphocyte Count , Cell Death/drug effects , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/pharmacology , Cytokines/blood , Exotoxins/pharmacology , Hyaluronan Receptors/analysis , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/pathology , Liver Failure/etiology , Liver Failure/prevention & control , Lymphocyte Count , Macrophages/physiology , Male , Mice , Mice, Inbred BALB C , Molecular Weight , NF-kappa B/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/physiology , Pseudomonas aeruginosa Exotoxin A
16.
Cancer Res ; 61(15): 5817-23, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11479221

ABSTRACT

Apoptosis is required for proper tissue homeostasis. Defects in apoptosis signaling pathways, thus, contribute to carcinogenesis and chemoresistance. A major goal in chemotherapy is, therefore, to find cytotoxic agents that restore the ability of tumor cells to undergo apoptosis. We show here that the sesquiterpene lactone helenalin (10-50 microM) induces apoptosis in leukemia Jurkat T cells even if they lack the CD95 death receptor or overexpress the antiapoptotic proteins Bcl-x(L) or Bcl-2. Activated peripheral blood mononuclear cells, however, are not affected (10-50 microM helenalin). Helenalin led to a time-dependent (0-24 h) cleavage of the specific caspase-3-like substrate Asp-Glu-Val-Asp-7-amino-4-trifluoromethylcoumarin as well as to the proteolytic processing of procaspase-3 and -8. Caspase activation was a necessary requirement for apoptosis because the broad-spectrum caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone (zVAD-fmk, 50 microM) completely abrogated helenalin-induced DNA fragmentation as well as phosphatidylserin translocation. Although the initiator caspase-8 was activated, the helenalin-induced signaling pathway did not require the CD95 death receptor as shown using cells without or with an antibody (ZB4)-blocked CD95 receptor. Helenalin also did not induce CD95 or CD95-ligand expression. On the other hand, helenalin was found to induce the release of cytochrome c from mitochondria that was not inhibited by the caspase inhibitor zVAD-fmk, which indicated that cytochrome c release precedes caspase activation. Cytochrome c release was accompanied by dissipation of the mitochondrial transmembrane potential (DeltaPsi(m)), which was partly inhibited by zVAD-fmk, which suggests that caspases are involved in loss of DeltaPsi(m). Most importantly, overexpression of the mitochondria protecting proteins Bcl-x(L) or Bcl-2 failed to confer resistance to helenalin-induced apoptosis, although the data presented here suggest that helenalin induces a mitochondria-dependent pathway. Thus, helenalin is a promising experimental cytotoxic agent that possibly points to new strategies to overcome apoptosis resistance attributable to overexpression of antiapoptotic Bcl-2 proteins.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Jurkat Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Sesquiterpenes/pharmacology , fas Receptor/physiology , Apoptosis/physiology , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Enzyme Activation , Enzyme Induction/drug effects , Fas Ligand Protein , Humans , Jurkat Cells/metabolism , Jurkat Cells/pathology , Matrix Metalloproteinases/biosynthesis , Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Sesquiterpenes, Guaiane , bcl-X Protein
17.
Planta Med ; 67(6): 557-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509981

ABSTRACT

Cytotoxicity is a well characterized property of sesquiterpene lactones. In the present study, the question was addressed whether sesquiterpene lactones mediate their cytotoxic effect by triggering apoptosis. Four compounds, ambrosin, alantolactone, hymenin and helenalin were shown to induce apoptosis in Jurkat leukemia T cells as judged by cell morphology, the appearance of apoptotic nuclei as well as the translocation of phosphatidylserine to the outer surface of the cell membrane.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Asteraceae/chemistry , Lactones/pharmacology , Sesquiterpenes/pharmacology , T-Lymphocytes/drug effects , Antineoplastic Agents, Phytogenic/chemistry , Cell Survival , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Jurkat Cells , Lactones/chemistry , Necrosis , Sesquiterpenes/chemistry
18.
Biochem Pharmacol ; 61(5): 587-93, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11239502

ABSTRACT

The inducible isoform of cyclooxygenase (COX-2) is implicated in the pathogenesis of various inflammatory diseases as well as in carcinogenesis, especially of gastrointestinal tumors. Epidemiological as well as experimental data support a role for constituents of allium vegetables, such as garlic and onions, in the prevention of gastrointestinal cancer. Therefore, the aim of the present study was to examine whether the garlic-derived natural product ajoene interferes with the COX-2 pathway by using lipopolysaccharide (LPS)-activated RAW 264.7 cells as in vitro model. Ajoene was shown to dose-dependently inhibit the release of LPS (1 microg/mL)-induced prostaglandin E(2) in RAW 264.7 macrophages (IC(50) value: 2.4 microM). This effect was found to be due to an inhibition of COX-2 enzyme activity by ajoene (IC(50) value: 3.4 microM). Ajoene did not reduce COX-2 expression, but rather increased LPS-induced COX-2 protein and mRNA expression compared to LPS-stimulated cells only. In the absence of LPS, however, ajoene was unable to induce COX-2. The non-steroidal anti-inflammatory drug indomethacin was shown to act similarly in LPS-activated RAW 264.7 cells. These data suggest that ajoene works by a mechanism of action similar to that attributed to non-steroidal anti-inflammatory drugs. This finding may add a novel aspect to the biological profile of the garlic-derived natural product ajoene which might be important for understanding the usefulness of garlic for chemoprevention of gastrointestinal carcinomas.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Disulfides/pharmacology , Isoenzymes/metabolism , Macrophages/drug effects , Plant Extracts/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Biological Factors/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2 , Dinoprostone/metabolism , Isoenzymes/drug effects , Isoenzymes/genetics , Lipopolysaccharides , Macrophage Activation/drug effects , Macrophages/enzymology , Macrophages/pathology , Mice , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Sulfoxides
19.
Immunol Cell Biol ; 79(1): 11-7, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11168617

ABSTRACT

Atrial natriuretic peptide (ANP) attenuates LPS-induced inducible nitric oxide synthase (iNOS) expression in murine macrophages by destabilizing iNOS mRNA. Because elevated intracellular free Ca2+ levels [Ca2+]i reduce iNOS mRNA stability, the aim of the present study was to determine whether inhibition of iNOS by ANP is due to alterations in intracellular calcium. As determined by fluorescence photometry, ANP (10(-7) and 10(-6) mol/L) was shown to elevate intracellular calcium levels in bone marrow-derived macrophages. This effect seemed to be mediated via the guanylate cyclase-coupled A receptor, because dibutyryl-cGMP mimicked and the A-receptor antagonist HS-142-1 partially abrogated the effect of ANP. Because the Ca2+ increase was also observed in Ca2+-free buffer, it is suggested that the liberation of intracellular calcium pools contributes to the elevation of [Ca2+]i by ANP. The B-receptor ligand C-type natriuretic peptide (CNP), which does not alter iNOS expression, had no effect on [Ca2+]i. The Ca2+-ionophore 4-Br-A23187 and thapsigargin, a compound known to liberate Ca2+ from intracellular stores, were further demonstrated to reduce LPS-induced NO production in macrophages (Griess assay), confirming a functional link for elevated [Ca2+]i and iNOS inhibition. These effects were abrogated by coincubation with extra- as well as intracellular Ca2+ chelators (EGTA, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA)). The inhibitory effect of ANP on NO production was also abrogated by Ca2+ chelation. These findings support a causal relationship between reduced iNOS induction and elevation of [Ca2+]i. Taken together, the data indicate that intracellular Ca2+ elevation by ANP is involved in the inhibition of LPS-induced nitric oxide production in macrophages.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Calcimycin/analogs & derivatives , Calcium/metabolism , Macrophages/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/biosynthesis , Aniline Compounds/metabolism , Animals , Atrial Natriuretic Factor/antagonists & inhibitors , Calcimycin/pharmacology , Cells, Cultured , Dibutyryl Cyclic GMP/pharmacology , Fluorescent Dyes/metabolism , Guanylate Cyclase/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Macrophages/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Polysaccharides/pharmacology , Receptors, Atrial Natriuretic Factor/antagonists & inhibitors , Thapsigargin/pharmacology , Xanthenes/metabolism
20.
Ann Rheum Dis ; 60 Suppl 3: iii68-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11890659

ABSTRACT

The atrial natriuretic peptide (ANP), a member of the natriuretic peptide family, is a cardiovascular hormone which possesses well defined natriuretic, diuretic, and vasodilating properties. Most of the biological effects of ANP aremediated through its guanylyl cyclase coupled A receptor. Because ANP and its receptors have been shown to be expressed and differentially regulated in the immune system, it has been suggested that ANP has an immunomodulatory potency. Much investigation of the effects of ANP on the activation of macrophages has been carried out. ANP was shown to inhibit the lipopolysaccharide (LPS)-induced expression of inducible nitric oxide synthase (iNOS) in macrophages in an autocrine fashion. ANP in this context was shown to reduce significantly the activation of NF-kappaB and to destabilise iNOS mRNA. ANP, furthermore, can significantly reduce the LPS-induced secretion of tumour necrosis factor alpha (TNFalpha) in macrophages. The relevance of these findings on a regulatory role for ANP on TNFalpha in humans was shown by the fact that ANP significantly reduces the release of TNFalpha in whole human blood. It was furthermore shown to attenuate the release of interleukin 1beta (IL1beta). Interestingly, ANP did not affect the secretion of the anti-inflammatory cytokines IL10 and IL1 receptor antagonist (IL1ra). In summary, ANP was shown to reduce the secretion of inflammatory mediators in macrophages. Therefore, this cardiovascular hormone may possess anti-inflammatory potential.


Subject(s)
Atrial Natriuretic Factor/physiology , Autocrine Communication/physiology , Inflammation Mediators/metabolism , Macrophages/immunology , Rheumatic Diseases/immunology , Humans , Macrophage Activation/physiology , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...