Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 21(6): 859-870, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35405736

ABSTRACT

Radiotherapy and chemical DNA-damaging agents are among the most widely used classes of cancer therapeutics today. Double-strand breaks (DSB) induced by many of these treatments are lethal to cancer cells if left unrepaired. Ataxia telangiectasia-mutated (ATM) kinase plays a key role in the DNA damage response by driving DSB repair and cell-cycle checkpoints to protect cancer cells. Inhibitors of ATM catalytic activity have been shown to suppress DSB DNA repair, block checkpoint controls and enhance the therapeutic effect of radiotherapy and other DSB-inducing modalities. Here, we describe the pharmacological activities of two highly potent and selective ATM inhibitors from a new chemical class, M3541 and M4076. In biochemical assays, they inhibited ATM kinase activity with a sub-nanomolar potency and showed remarkable selectivity against other protein kinases. In cancer cells, the ATM inhibitors suppressed DSB repair, clonogenic cancer cell growth, and potentiated antitumor activity of ionizing radiation in cancer cell lines. Oral administration of M3541 and M4076 to immunodeficient mice bearing human tumor xenografts with a clinically relevant radiotherapy regimen strongly enhanced the antitumor activity, leading to complete tumor regressions. The efficacy correlated with the inhibition of ATM activity and modulation of its downstream targets in the xenograft tissues. In vitro and in vivo experiments demonstrated strong combination potential with PARP and topoisomerase I inhibitors. M4076 is currently under clinical investigation.


Subject(s)
Ataxia Telangiectasia , Neoplasms , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Repair , Humans , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology
2.
J Med Chem ; 65(2): 1265-1282, 2022 01 27.
Article in English | MEDLINE | ID: mdl-35081715

ABSTRACT

NIMA-related kinase 1 (Nek1) has lately garnered attention for its widespread function in ciliogenesis, apoptosis, and the DNA-damage response. Despite its involvement in various diseases and its potential as a cancer drug target, no directed medicinal chemistry efforts toward inhibitors against this dark kinase are published. Here, we report the structure-guided design of a potent small-molecule Nek1 inhibitor, starting from a scaffold identified by kinase cross-screening analysis. Seven lead compounds were identified in silico and evaluated for their inhibitory activity. The top compound, 10f, was further profiled for efficacy, toxicity, and bioavailability in a zebrafish polycystic kidney disease model. Administration of 10f caused the expansion of fluorescence-labeled proximal convoluted tubules, supporting our hypothesis that Nek1-inhibition causes cystic kidneys in zebrafish embryos. Compound 10f displayed insignificant inhibition in 48 of 50 kinases in a selectivity test panel. The findings provide a powerful tool to further elucidate the function and pharmacology of this neglected kinase.


Subject(s)
Drug Design , Embryo, Nonmammalian/drug effects , NIMA-Related Kinase 1/antagonists & inhibitors , Polycystic Kidney Diseases/drug therapy , Pronephros/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Embryo, Nonmammalian/enzymology , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/pathology , Pronephros/embryology , Pronephros/enzymology , Zebrafish
3.
Mol Cancer Ther ; 19(5): 1091-1101, 2020 05.
Article in English | MEDLINE | ID: mdl-32220971

ABSTRACT

Physical and chemical DNA-damaging agents are used widely in the treatment of cancer. Double-strand break (DSB) lesions in DNA are the most deleterious form of damage and, if left unrepaired, can effectively kill cancer cells. DNA-dependent protein kinase (DNA-PK) is a critical component of nonhomologous end joining (NHEJ), one of the two major pathways for DSB repair. Although DNA-PK has been considered an attractive target for cancer therapy, the development of pharmacologic DNA-PK inhibitors for clinical use has been lagging. Here, we report the discovery and characterization of a potent, selective, and orally bioavailable DNA-PK inhibitor, M3814 (peposertib), and provide in vivo proof of principle for DNA-PK inhibition as a novel approach to combination radiotherapy. M3814 potently inhibits DNA-PK catalytic activity and sensitizes multiple cancer cell lines to ionizing radiation (IR) and DSB-inducing agents. Inhibition of DNA-PK autophosphorylation in cancer cells or xenograft tumors led to an increased number of persistent DSBs. Oral administration of M3814 to two xenograft models of human cancer, using a clinically established 6-week fractionated radiation schedule, strongly potentiated the antitumor activity of IR and led to complete tumor regression at nontoxic doses. Our results strongly support DNA-PK inhibition as a novel approach for the combination radiotherapy of cancer. M3814 is currently under investigation in combination with radiotherapy in clinical trials.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , DNA-Activated Protein Kinase/antagonists & inhibitors , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Quinazolines/pharmacology , Radiation, Ionizing , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
SLAS Discov ; 22(10): 1203-1210, 2017 12.
Article in English | MEDLINE | ID: mdl-28820955

ABSTRACT

Mass spectrometry (MS) is known for its label-free detection of substrates and products from a variety of enzyme reactions. Recent hardware improvements have increased interest in the use of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS for high-throughput drug discovery. Despite interest in this technology, several challenges remain and must be overcome before MALDI-MS can be integrated as an automated "in-line reader" for high-throughput drug discovery. Two such hurdles include in situ sample processing and deposition, as well as integration of MALDI-MS for enzymatic screening assays that usually contain high levels of MS-incompatible components. Here we adapt our c-MET kinase assay to optimize for MALDI-MS compatibility and test its feasibility for compound screening. The pros and cons of the Echo (Labcyte) as a transfer system for in situ MALDI-MS sample preparation are discussed. We demonstrate that this method generates robust data in a 1536-grid format. We use the MALDI-MS to directly measure the ratio of c-MET substrate and phosphorylated product to acquire IC50 curves and demonstrate that the pharmacology is unaffected. The resulting IC50 values correlate well between the common label-based capillary electrophoresis and the label-free MALDI-MS detection method. We predict that label-free MALDI-MS-based high-throughput screening will become increasingly important and more widely used for drug discovery.


Subject(s)
High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-met/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Inhibitory Concentration 50 , Substrate Specificity
5.
Bioorg Med Chem Lett ; 25(7): 1597-602, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25736998

ABSTRACT

In a high-throughput screening campaign for c-Met kinase inhibitors, a thiadiazinone derivative with a carbamate group was identified as a potent in vitro inhibitor. Subsequent optimization guided by c-Met-inhibitor X-ray structures furnished new compound classes with excellent in vitro and in vivo profiles. The thiadiazinone ring of the HTS hit was first replaced by a pyridazinone followed by an exchange of the carbamate hinge binder with a 1,5-disubstituted pyrimidine. Finally an optimized compound, 22 (MSC2156119), with excellent in vitro potency, high kinase selectivity, long half-life after oral administration and in vivo anti-tumor efficacy at low doses, was selected as a candidate for clinical development.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridazines/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/metabolism , Pyridazines/chemical synthesis , Pyridazines/chemistry , Structure-Activity Relationship
6.
Clin Cancer Res ; 19(11): 2941-51, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23553846

ABSTRACT

PURPOSE: The mesenchymal-epithelial transition factor (c-Met) receptor, also known as hepatocyte growth factor receptor (HGFR), controls morphogenesis, a process that is physiologically required for embryonic development and tissue repair. Aberrant c-Met activation is associated with a variety of human malignancies including cancers of the lung, kidney, stomach, liver, and brain. In this study, we investigated the properties of two novel compounds developed to selectively inhibit the c-Met receptor in antitumor therapeutic interventions. EXPERIMENTAL DESIGN: The pharmacologic properties, c-Met inhibitory activity, and antitumor effects of EMD 1214063 and EMD 1204831 were investigated in vitro and in vivo, using human cancer cell lines and mouse xenograft models. RESULTS: EMD 1214063 and EMD 1204831 selectively suppressed the c-Met receptor tyrosine kinase activity. Their inhibitory activity was potent [inhibitory 50% concentration (IC50), 3 nmol/L and 9 nmol/L, respectively] and highly selective, when compared with their effect on a panel of 242 human kinases. Both EMD 1214063 and EMD 1204831 inhibited c-Met phosphorylation and downstream signaling in a dose-dependent fashion, but differed in the duration of their inhibitory activity. In murine xenograft models, both compounds induced regression of human tumors, regardless of whether c-Met activation was HGF dependent or independent. Both drugs were well tolerated and induced no substantial weight loss after more than 3 weeks of treatment. CONCLUSIONS: Our results indicate selective c-Met inhibition by EMD 1214063 and EMD 1204831 and strongly support clinical testing of these compounds in the context of molecularly targeted anticancer strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Morpholines/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridazines/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridazines/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Biochem J ; 427(1): 151-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20100168

ABSTRACT

Vesicular V-ATPase (V-type H+-ATPase) and the plasma membrane-bound Na+/K+-ATPase are essential for the cycling of neurotransmitters at the synapse, but direct functional studies on their action in native surroundings are limited due to the poor accessibility via standard electrophysiological equipment. We performed SSM (solid supported membrane)-based electrophysiological analyses of synaptic vesicles and plasma membranes prepared from rat brains by sucrose-gradient fractionation. Acidification experiments revealed V-ATPase activity in fractions containing the vesicles but not in the plasma membrane fractions. For the SSM-based electrical measurements, the ATPases were activated by ATP concentration jumps. In vesicles, ATP-induced currents were inhibited by the V-ATPase-specific inhibitor BafA1 (bafilomycin A1) and by DIDS (4,4'-di-isothiocyanostilbene-2,2'-disulfonate). In plasma membranes, the currents were inhibited by the Na+/K+-ATPase inhibitor digitoxigenin. The distribution of the V-ATPase- and Na+/K+-ATPase-specific currents correlated with the distribution of vesicles and plasma membranes in the sucrose gradient. V-ATPase-specific currents depended on ATP with a K0.5 of 51+/-7 microM and were inhibited by ADP in a negatively co-operative manner with an IC50 of 1.2+/-0.6 microM. Activation of V-ATPase had stimulating effects on the chloride conductance in the vesicles. Low micromolar concentrations of DIDS fully inhibited the V-ATPase activity, whereas the chloride conductance was only partially affected. In contrast, NPPB [5-nitro-2-(3-phenylpropylamino)-benzoic acid] inhibited the chloride conductance but not the V-ATPase. The results presented describe electrical characteristics of synaptic V-ATPase and Na+/K+-ATPase in their native surroundings, and demonstrate the feasibility of the method for electrophysiological studies of transport proteins in native intracellular compartments and plasma membranes.


Subject(s)
Brain/enzymology , Cell Membrane/enzymology , Electrophysiology , Sodium-Potassium-Exchanging ATPase/metabolism , Synaptic Membranes/enzymology , Synaptic Vesicles/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Macrolides/pharmacology , Membrane Potentials/drug effects , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
8.
Brain Res ; 1241: 42-55, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18817758

ABSTRACT

The sphenopalatine ganglia (SPG) receive their preganglionic innervation from the ventro-lateral reticular formation and nuclei of the caudal pons, and are involved in parasympathetic control of cranial glandular and vascular components including the blood supply to specific brain areas. In 53% of all SPG neurons, a particular member (MOL2.3) of the odorant receptor superfamily is co-expressed with green fluorescent protein (GFP) in MOL2.3 transgenic mouse pups. Choline acetyltransferase and vesicular acetylcholine transporter (VAChT) could be demonstrated in 90% of the GFP-positive, and 60% of the GFP-negative cells, these cells thus representing cholinergic neurons. Some 50% of all SPG neurons were nitrergic at a high rate of VAChT co-expression, the majority of them being GFP-positive. Most SPG neurons received cholinergic innervation as demonstrated by perineuronal VAChT immunoreactive nerve terminals. To characterize cholinergic signal transduction in SPG neurons, calcium imaging experiments were performed in a SPG primary culture system containing GFP-positive and -negative neurons. Ganglionic neurons could repeatedly be activated by cholinergic stimulation in a dose-dependent manner, with calcium entering all cells from the extracellular compartment. Stimulation with specific agonists supported prevalence of nicotinic cholinergic receptors (nAChRs). Inhibition of cholinergically induced intracellular calcium signalling by various omega-conotoxins indicated functional expression of alpha 3 beta 4 and alpha 7 nAChR subtypes in murine SPG cells, which could be supported by RT-PCR analysis of the neonatal mouse SPG. With regard to secondary cholinergic activation, L- but not N-subtype voltage-gated calcium channels might represent a prime target. Nicotinic signal transduction did not prove to be different in GFP-positive as compared to-negative murine SPG neurons.


Subject(s)
Acetylcholine/metabolism , Calcium Signaling/physiology , Cholinergic Fibers/metabolism , Ganglia, Parasympathetic/metabolism , Neurons/metabolism , Receptors, Nicotinic/metabolism , Animals , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/drug effects , Cholinergic Fibers/ultrastructure , Dose-Response Relationship, Drug , Ganglia, Parasympathetic/cytology , Ganglia, Parasympathetic/drug effects , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Nicotinic Agonists/pharmacology , Nitrergic Neurons/cytology , Nitrergic Neurons/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Receptors, Nicotinic/drug effects , Receptors, Odorant/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Vesicular Acetylcholine Transport Proteins/metabolism , alpha7 Nicotinic Acetylcholine Receptor , omega-Conotoxins/pharmacology
9.
J Cell Biol ; 172(2): 221-31, 2006 Jan 16.
Article in English | MEDLINE | ID: mdl-16418534

ABSTRACT

Mammalian Staufen2 (Stau2) is a member of the double-stranded RNA-binding protein family. Its expression is largely restricted to the brain. It is thought to play a role in the delivery of RNA to dendrites of polarized neurons. To investigate the function of Stau2 in mature neurons, we interfered with Stau2 expression by RNA interference (RNAi). Mature neurons lacking Stau2 displayed a significant reduction in the number of dendritic spines and an increase in filopodia-like structures. The number of PSD95-positive synapses and miniature excitatory postsynaptic currents were markedly reduced in Stau2 down-regulated neurons. Akin effects were caused by overexpression of dominant-negative Stau2. The observed phenotype could be rescued by overexpression of two RNAi cleavage-resistant Stau2 isoforms. In situ hybridization revealed reduced expression levels of beta-actin mRNA and fewer dendritic beta-actin mRNPs in Stau2 down-regulated neurons. Thus, our data suggest an important role for Stau2 in the formation and maintenance of dendritic spines of hippocampal neurons.


Subject(s)
Brain/metabolism , Dendrites , Neurons/cytology , Protein Isoforms/metabolism , RNA-Binding Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Dendrites/metabolism , Dendrites/ultrastructure , Disks Large Homolog 4 Protein , Down-Regulation , Excitatory Postsynaptic Potentials/physiology , HeLa Cells , Hippocampus/cytology , Humans , In Situ Hybridization , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neurons/physiology , Patch-Clamp Techniques , Protein Isoforms/genetics , RNA Interference , RNA-Binding Proteins/genetics , Rats , Synapses/metabolism
10.
Assay Drug Dev Technol ; 2(5): 515-24, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15671649

ABSTRACT

As numerous diseases have been shown to be related to dysfunction of ion channels and neurotransmitter receptors and to affect regulatory pathways, ion channels have attracted increasing attention as a target class for drug discovery. The concomitant demand of the pharmaceutical industry for adequate electrophysiological methods to investigate drug effects on specific ion channels in secondary and safety screening has resulted in the development of electrophysiological instrumentation that allows automated monitoring of ion channel function with a higher throughput. Here we tested a fully automated screening system based on the Xenopus laevis oocyte expression system. We addressed the questions of data quality and reproducibility obtained by automated oocyte injection and two-electrode voltage-clamp (TEVC) recording using the Roboocyte (Multi Channel Systems GmbH, Reutlingen, Germany) technology compared to conventional oocyte recording. A gamma-aminobutyric acid (GABA)A-receptor subtype (alpha(1)beta(2)) was chosen as an example for a ligand-gated ion channel, and the slowly activating potassium current I(Ks) as a voltage-activated ion channel. Oocytes were injected with cDNA or cRNA via the Roboocyte injection stage. Ion channel currents were successfully recorded after 2-7 days in about 40% of the oocytes injected with GABA(A) receptor cDNA, and after 2-4 days in about 60% of the oocytes injected with KCNE1 cRNA. EC(50) values for the GABA(A) receptor and IC(50) values for blockers of I(Ks) were comparable to values obtained with conventional TEVC recording techniques. In conclusion, our results show that the Roboocyte is a valuable automated tool for oocyte injection and TEVC recording that can be used in drug screening and target validation to enhance the number of compounds and oocytes tested per day.


Subject(s)
Drug Delivery Systems/methods , Ion Channels/physiology , Robotics/methods , Animals , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Injections , Oocytes/metabolism , Receptors, GABA-A/administration & dosage , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Robotics/instrumentation , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
11.
J Appl Physiol (1985) ; 94(4): 1395-402, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12482772

ABSTRACT

In guinea pigs, dose-dependent febrile responses can be induced by injection of a high (100 micro g/kg) or low (10 micro g/kg) dose of bacterial lipopolysaccharide (LPS) into artificial subcutaneously implanted Teflon chambers. In this fever model, LPS does not enter the systemic circulation from the site of localized tissue inflammation in considerable amounts but causes a local induction of the proinflammatory cytokines tumor necrosis factor (TNF) and interleukin-6 (IL-6), which can be measured in lavage fluid collected from the chamber area. Only in response to the high LPS dose, small traces of TNF are measurable in blood plasma. A moderate increase of circulating IL-6 occurs in response to administration of both LPS doses. To investigate the putative roles of TNF and prostaglandins in this fever model, a neutralizing TNF binding protein (TNF-bp) or a nonselective inhibitor of cyclooxygenases (diclofenac) was injected along with the high or low dose of LPS into the subcutaneous chamber. In control groups, both doses of LPS were administered into the chamber along with the respective vehicles for the applied drugs. The fever response to the high LPS dose remained unimpaired by treatment with TNF-bp despite an effective neutralization of bioactive TNF in the inflamed tissue area. In response to the low LPS dose, there was an accelerated defervescence under the influence of TNF-bp. Blockade of prostaglandin formation with diclofenac completely abolished fever in response to both LPS doses. In conclusion, prostaglandins seem to be essential components for the manifestation of fever in this model.


Subject(s)
Cytokines/metabolism , Drug Eruptions/complications , Drug Eruptions/metabolism , Fever/etiology , Prostaglandins/metabolism , Receptors, Tumor Necrosis Factor , Animals , Carrier Proteins/pharmacology , Cyclooxygenase Inhibitors/administration & dosage , Diclofenac/administration & dosage , Drug Combinations , Fever/physiopathology , Guinea Pigs , Injections, Subcutaneous , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/blood , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor Decoy Receptors
12.
J Appl Physiol (1985) ; 92(6): 2657-66, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12015387

ABSTRACT

Interleukin-6 (IL-6) is regarded as an endogenous mediator of lipopolysaccharide (LPS)-induced fever. IL-6 is thought to act on the brain at sites that lack a blood-brain barrier, the circumventricular organs (CVOs). Cells that are activated by IL-6 respond with nuclear translocation of the signal transducer and activator of transcription 3 molecule (STAT3) and can be detected by immunohistochemistry. We investigated whether the LPS-induced release of IL-6 into the systemic circulation was accompanied by a nuclear STAT3 translocation within the sensory CVOs. Treatment with LPS (100 microg/kg) led to a slight (1 h) and then a strong increase (2-8 h) in plasma IL-6 levels, which started to decline at the end of the febrile response. Administration of both pyrogens LPS and IL-6 (45 microg/kg) induced a febrile response with IL-6, causing a rather moderate fever compared with the LPS-induced fever. Nuclear STAT3 translocation in response to LPS was observed within the vascular organ of the lamina terminalis (OVLT) and the subfornical organ (SFO) 2 h after LPS treatment. To investigate whether this effect was mediated by IL-6, the cytokine itself was systemically applied and indeed an identical pattern of nuclear STAT3 translocation was observed. However, nuclear STAT3 translocation already occurred 1 h after IL-6 application and proved to be less effective compared with LPS treatment when analyzing OVLT and SFO cell numbers that showed nuclear STAT3 immunoreactivity after the respective pyrogen treatment. Our observations represent the first molecular evidence for an IL-6-induced STAT3-mediated genomic activation of OVLT and SFO cells and support the proposed role of these brain areas as sensory structures for humoral signals created by the activated immune system and resulting in the generation of fever.


Subject(s)
Brain/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Fever/metabolism , Interleukin-6/physiology , Lipopolysaccharides/pharmacology , Trans-Activators/metabolism , Animals , Biological Transport/physiology , Cytoplasm/metabolism , Fourth Ventricle/metabolism , Hypothalamus/metabolism , Interleukin-6/pharmacology , Rats , Rats, Wistar , STAT3 Transcription Factor , Subfornical Organ/metabolism , Tissue Distribution
13.
J Neurosci Res ; 68(2): 176-84, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11948663

ABSTRACT

Certain members of the olfactory receptor superfamily appear to be expressed not only in chemosensory neurons of the nasal epithelium. Analyzing the transgenic mouse line MOL2.3-IGITL, the olfactory receptor subtype MOL2.3 was found to be expressed in distinct subpopulations of cells within a cranial, a cervical as well as within a thoracic ganglion. By means of coexpressed markers, the axonal processes of MOL2.3 expressing cells could be visualized and thus the target tissues innervated by these ganglionic neurons identified. Stained fibers, but no stained cell bodies were visible in distinct head regions, notably in the lateral nasal gland and in the so-called Harderian gland; staining was also observed on distinct segments of blood vessels, especially within the tongue. In the thoracic region, the heart and a small segment of the aorta as well as a distinct population of lung alveoli were labeled by incoming blue fibers. Expression of MOL2.3 in cells of the autonomic nervous system supports the idea that at least some of the multiple olfactory receptor types serve functions others than odorant detection.


Subject(s)
Ganglia, Autonomic/metabolism , Receptors, Odorant/metabolism , Animals , Brain , Gene Expression , Mice , Mice, Transgenic/genetics , Neck/innervation , Nerve Fibers/metabolism , Receptors, Odorant/genetics , Thorax/innervation , Tissue Distribution
14.
Pflugers Arch ; 443(3): 411-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11810211

ABSTRACT

Peripheral inflammatory stimuli result in the modification of a number of vital brain-controlled functions including the thermoregulatory set-point (induction of fever) and the activity of the hypothalamic-pituitary-adrenal (HPA) axis. We addressed the question of whether both of these components of the acute-phase response are induced by a common signal pathway. For this purpose we recorded body temperature (by remote radio-telemetry), HPA axis activity (circulating concentrations of cortisol by radio-immunoassay) and levels of the pro-inflammatory cytokines tumour necrosis factor and interleukin-6 (TNF, IL-6, using specific bioassays) in six groups of guinea-pigs. The animals received intra-arterial injections of either 10 microg/kg lipopolysaccharide (LPS) plus saline, 10 microg/kg LPS plus 5 mg/kg meloxicam (an inhibitor of the inducible form of cyclooxygenase), 10 microg/kg LPS plus 5 mg/kg diclofenac (a non-selective cyclooxygenase inhibitor), saline plus solvent, saline plus 5 mg/kg meloxicam or saline plus 5 mg/kg diclofenac. Injection of the cyclooxygenase inhibitors per se had no influence on the investigated parameters. Injection of LPS alone resulted in a biphasic fever, a more than fivefold increase in circulating cortisol and pronounced induction of TNF and IL-6. Treatment with the cyclooxygenase inhibitors either attenuated (meloxicam) or abolished (diclofenac) LPS-induced fever, but had no effect on the LPS-induced rise of plasma cortisol or IL-6. Circulating levels of TNF, in response to LPS, were enhanced by meloxicam and diclofenac, reflecting the negative feedback control exerted by prostaglandins on cytokine (specifically TNF) formation. These results provide the first evidence that the prostaglandin-dependent inflammatory pathway for fever induction is distinct from the pathway of HPA axis activation since fever, but not circulating cortisol, was attenuated by an inhibition of prostaglandin formation.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Fever/metabolism , Hydrocortisone/blood , Interleukin-6/blood , Thiazines/pharmacology , Thiazoles/pharmacology , Animals , Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Diclofenac/pharmacology , Fever/chemically induced , Fever/immunology , Guinea Pigs , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/immunology , Lipopolysaccharides , Male , Meloxicam , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/immunology , Prostaglandins/physiology , Telemetry , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...