Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Biochem Pharmacol ; 73(8): 1157-71, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17288995

ABSTRACT

Brain histaminergic neurons play a prominent role in arousal and maintenance of wakefulness (W). H(3)-receptors control the activity of histaminergic neurons through presynaptic autoinhibition. The role of H(3)-receptor antagonists/inverse agonists (H(3)R-antagonists) in the potential therapy of vigilance deficiency and sleep-wake disorders were studied by assessing their effects on the mouse cortical EEG and sleep-wake cycle in comparison to modafinil and classical psychostimulants. The H(3)R-antagonists, thioperamide and ciproxifan increased W and cortical EEG fast rhythms and, like modafinil, but unlike amphetamine and caffeine, their waking effects were not accompanied by sleep rebound. Conversely, imetit (H(3)R-agonist) enhanced slow wave sleep and dose-dependently attenuated ciproxifan-induced W, indicating that the effects of both ligands involve H(3)-receptor mechanisms. Additional studies using knockout (KO) mice confirmed the essential role of H(3)-receptors and histamine-mediated transmission in the wake properties of H(3)R-antagonists. Thus ciproxifan produced no increase in W in either histidine-decarboxylase (HDC, histamine-synthesizing enzyme) or H(1)- or H(3)-receptor KO-mice whereas its waking effects persisted in H(2)-receptor KO-mice. These data validate the hypothesis that H(3)R-antagonists, through disinhibition of H(3)-autoreceptors, enhancing synaptic histamine that in turn activates postsynaptic H(1)-receptors promoting W. Interestingly amphetamine and modafinil, despite their potent arousal effects, appear unlikely to depend on histaminergic mechanism as their effects still occurred in HDC KO-mice. The present study thus distinguishes two classes of wake-improving agents: the first acting through non-histaminergic mechanisms and the second acting via histamine and supports brain H(3)-receptors as potentially novel therapeutic targets for vigilance and sleep-wake disorders.


Subject(s)
Benzhydryl Compounds/pharmacology , Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Histamine/metabolism , Receptors, Histamine H3/physiology , Animals , Benzhydryl Compounds/therapeutic use , Brain/drug effects , Brain/physiology , Electroencephalography/drug effects , Histamine Agonists/therapeutic use , Histamine Antagonists/therapeutic use , Mice , Mice, Knockout , Modafinil , Models, Animal , Sleep/drug effects , Sleep/physiology , Sleep Wake Disorders/drug therapy , Wakefulness/drug effects , Wakefulness/physiology
2.
J Neuropathol Exp Neurol ; 55(7): 850-60, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8965100

ABSTRACT

Calpain, a calcium-activated neutral protease family, has been implicated in the neuropathologic sequelae accompanying various neurological disorders. We have characterized the distribution and time course of calpain activation following brain injury in the rat, using a monoclonal antibody that recognizes calpain-generated breakdown products (BDPs) of spectrin. Adult male Sprague-Dawley rats received lateral fluid percussion brain injury of moderate severity (2.2-2.4 atm, n = 35) or served as controls (uninjured, n = 12). One group of animals (n = 21) were sacrificed at either 30 minutes (min), 1 day, or 3 days post-injury, and selected brain regions were prepared for Western blot analysis. The remaining animals (n = 26) were sacrificed at 90 min, 4 hours (h), 1 day, or 7 days post-injury, and immunohistochemistry was performed. Spectrin BDPs were found predominantly in the hemisphere ipsilateral to the injury site, located primarily in cortical and hippocampal regions which exhibit neuronal death. Calpain-mediated spectrin breakdown was detected at 90 min in dendrites and axons, and by 4 h in neuronal perikarya. By 1 day post-injury, cortical and hippocampal regions of calpain activation had increased in size. Delayed spectrin breakdown was observed in the thalamus, both at 3 days and 7 days after injury. These results suggest that calpain may play an important role in the neurodegenerative process following brain injury.


Subject(s)
Brain Injuries/metabolism , Calpain/metabolism , Spectrin/metabolism , Animals , Blotting, Western , Brain/metabolism , Enzyme Activation , Immunohistochemistry , Male , Rabbits , Rats , Rats, Sprague-Dawley , Time Factors , Tissue Distribution
3.
J Med Chem ; 40(23): 3820-8, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9371247

ABSTRACT

Calpain I, an intracellular cysteine protease, has been implicated in the neurodegeneration following an episode of stroke. In this paper, we report on a series of potent dipeptide fluoromethyl ketone inhibitors of recombinant human calpain I (rh calpain I). SAR studies revealed that while calpain I tolerates a variety of hydrophobic groups at the P1 site, Leu at P2 is preferred. However, the nature of the N-terminal capping group has a significant effect on the inhibitory activity of this series of compounds. Compound 4e [(1,2,3,4-tetrahydroisoquinolin-2-yl)carbonyl-Leu-D,L-Phe-CH2F+ ++], having a tetrahydroisoquinoline containing urea as the N-terminal capping group, is the most potent dipeptide fluoromethyl ketone inhibitor of calpain I (with a second-order rate constant for inactivation of 276,000 M-1 s-1) yet reported; tripeptide 4k (Cbz-Leu-Leu-D,L-Phe-CH2F) is equipotent. A number of compounds presented in this study displayed excellent selectivity for calpain I over cathepsins B and L, two related cysteine proteases. Compounds which exhibited good inhibitory activity in the assay against isolated rh calpain I also inhibited intracellular calpain I in a human cell line. Thus, in an intact cell assay, compounds 4e and 4k inhibited calpain I with IC50 values of 0.2 and 0.1 microM, respectively. Finally, we also disclose the first example of fluorination of a dipeptide enol silyl ether to generate the corresponding dipeptide fluoromethyl ketone.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Endopeptidases , Ketones/chemical synthesis , Ketones/pharmacology , Calpain/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cathepsin L , Cathepsins/antagonists & inhibitors , Cathepsins/metabolism , Cysteine Endopeptidases , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/pharmacology , Leukemia, T-Cell/enzymology , Recombinant Proteins/metabolism , Tumor Cells, Cultured/drug effects
4.
Neuroscience ; 92(4): 1425-42, 1999.
Article in English | MEDLINE | ID: mdl-10426497

ABSTRACT

Activation of a member of the caspase family of cysteine proteases is thought to be required for the execution of apoptosis in neurons and other cell types. We describe here an antibody (Ab127) reactive with a neoantigenic site on caspase substrate proteins degraded during apoptosis, and its characterization as a biochemical and histochemical probe for apoptosis-associated proteolysis in growth factor-deprived neural cells in vitro and the developing postnatal rat brain. Neuronally differentiated PC12 cells became strongly Ab127 immunoreactive only during apoptosis following nerve growth factor withdrawal. Apoptosis-associated caspase proteolysis was detectable on western blots as markedly increased immunoreactivity of a approximately 46,000 mol. wt polypeptide, a product also generated by caspase-3 treatment of cell-free extracts. In the postnatal rat brain, intense immunoreactivity indicative of caspase activation was exhibited by small proportions of neurons and glia in distinct regional and temporal patterns. The degenerating nature of these cells was confirmed by their argyrophilia, cytoplasmic immunoreactivity for c-jun and fragmented processes. Combined immunofluorescence and Hoechst 33342 staining demonstrated that cells immunopositive for caspase activation have apoptotic nuclear morphologies. Caspase proteolysis was observed throughout the neuraxis in a minority of progenitor cells in germinal zones, postmitotic neurons in the parenchyma, and glia in the corpus callosum and other white matter tracts, but was observed rarely in the adult brain. These data characterize a new approach for evaluating apoptosis in physiological and pathological neurodegeneration, and demonstrate that caspase-associated apoptosis is a widespread mechanism for the programmed death of neurons and glia in the postnatal rat brain.


Subject(s)
Animals, Newborn/physiology , Apoptosis/physiology , Brain/cytology , Brain/enzymology , Caspases/metabolism , Caspases/physiology , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Neurons/physiology , Animals , Antibodies, Monoclonal , Cell Nucleus/immunology , Cell Nucleus/ultrastructure , Cells, Cultured , Fluorescent Antibody Technique, Direct , Immunoblotting , Immunohistochemistry , PC12 Cells , Proto-Oncogene Proteins c-jun/biosynthesis , Rats , Rats, Sprague-Dawley
5.
J Neurosci Methods ; 50(2): 205-16, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8107502

ABSTRACT

We report the development and characterization of a rapid fluorometric microassay suitable for quantifying neuronal cell survival. The method can be used in two formats: (1) a time course analysis of survival response or (2) as a simple endpoint assay for the assessment of neuronal survival promoted by a variety of reagents. The assay uses calcein AM, a non-fluorescent, electrically neutral, non-polar analogue of fluorescein diacetate, which passively crosses cell membranes and is cleaved to a fluorescent derivative by non-specific intracellular esterases. Once cleaved in viable cells, the resultant fluorescent salts are retained by intact cell membranes. The relative number of viable cells under various conditions can be quantified by measuring the emitted fluorescence. Described herein are the conditions that allow the determination of low viable neuronal cell numbers (10(2)-10(3) cells/cm2).


Subject(s)
Cell Survival , Cerebral Cortex/cytology , Neurons/cytology , Animals , Animals, Newborn , Cell Survival/drug effects , Cells, Cultured , Culture Techniques/methods , Fetus , Fluoresceins , Humans , Indicators and Reagents , Insulin-Like Growth Factor I/pharmacology , Microscopy, Phase-Contrast/methods , Neurons/drug effects , Rats , Recombinant Proteins/pharmacology , Retina/cytology , Spectrometry, Fluorescence/methods , Spinal Cord/cytology
9.
Biochem Biophys Res Commun ; 248(2): 293-6, 1998 Jul 20.
Article in English | MEDLINE | ID: mdl-9675129

ABSTRACT

We have developed a sensitive and continuously recording fluorogenic assay for the thiol protease calpain. This assay uses the dipeptide substrate Suc-Leu-Tyr-4-Methoxy-2-Naphthylamine (Suc-LY-MNA) in Tris buffer (pH 7.5) in the presence of 0.1% CHAPS. The assay is linear over a wide range of enzyme concentration and is capable of detecting 10 picomolar calpain making it more sensitive than any previously published method. Moreover, this assay gives a rate that is linear for over ten minutes making it useful for mechanistic studies of inhibitors. This assay can be easily adapted to a 96-well plate format facilitating the large scale screening of inhibitors.


Subject(s)
Calpain/analysis , Erythrocytes/enzymology , Cholic Acids/pharmacology , Detergents/pharmacology , Dipeptides/metabolism , Enzyme Inhibitors/analysis , Fluorometry , Humans , Kinetics , Naphthalenes/metabolism , Recombinant Proteins/metabolism , Sensitivity and Specificity , Substrate Specificity
10.
J Neurochem ; 77(3): 849-63, 2001 May.
Article in English | MEDLINE | ID: mdl-11331414

ABSTRACT

Although the mechanism of neuronal death in Alzheimer's disease (AD) has yet to be elucidated, a putative role for c-jun in this process has emerged. Thus, it was of interest to delineate signal transduction pathway(s) which regulate the transcriptional activity of c-jun, and relate these to alternate gene inductions and biochemical processes associated with beta-amyloid (Abeta) treatment. In this regard, the survival promoting activity of CEP-1347, an inhibitor of the stress-activated/c-jun N-terminal (SAPK/JNK) kinase pathway, was evaluated against Abeta-induced cortical neuron death in vitro. Moreover, CEP-1347 was used as a pharmacologic probe to associate multiple biochemical events with Abeta-induced activation of the SAPK/JNK pathway. CEP-1347 promoted survival and blocked Abeta-induced activation of JNK kinase (MKK4, also known as MEK-4, JNKK and SEK1) as well as other downstream events associated with JNK pathway activation. CEP-1347 also blocked Abeta-induction of cyclin D1 and DP5 genes and blocked Abeta-induced increases in cytoplasmic cytochrome c, caspase 3-like activity and calpain activation. The critical time window for cell death blockade by CEP-1347 resided within the peak of Abeta-induced MKK4 activation, thus defining this point as the most upstream event correlated to its survival-promoting activity. Together, these data link the SAPK/JNK pathway and multiple biochemical events associated with Abeta-induced neuronal death and further delineate the point of CEP-1347 interception within this signal transduction cascade.


Subject(s)
Amyloid beta-Peptides/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurons/physiology , Amyloid beta-Peptides/genetics , Animals , Calpain/metabolism , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cytochrome c Group/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme Activation/drug effects , Female , Gene Expression/drug effects , L-Lactate Dehydrogenase/metabolism , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/ultrastructure , Pregnancy , Rats , Rats, Sprague-Dawley
11.
Biochem J ; 314 ( Pt 2): 511-9, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8670065

ABSTRACT

Calpain I is a heterodimeric protein that is part of a family of calcium-activated intracellular cysteine proteases presumed to play a role in mediating signals transduced by calcium. Expression of bioactive recombinant human calpain I has been achieved using the baculovirus expression system, by either co-infection with two viruses, each expressing one of the subunits, or infection with a single virus containing both subunits. The approximately 80 kDa catalytic subunit exhibited calcium-dependent proteolytic activity when expressed alone or with the approximately 30 kDa regulatory subunit. Baculoviral recombinant calpain I appeared fully active in that the catalytic subunit in unpurified cell extracts exhibited calcium-dependent autocatalytic cleavage at the correct locus. The amount of approximately 80 kDa subunit accumulated at steady state was greatly increased by co-expression of the approximately 30 kDa subunit, suggesting a possible role for enzyme stabilization by the latter subunit. The recombinant human calpain I was purified to near homogeneity and compared with purified native human erythrocyte calpain I. The recombinant and native enzymes had equivalent inhibition constants for structurally diverse calpain inhibitors, identical calcium activation profiles, and similar specific activities, demonstrating the suitability of using the recombinant protein for studies of the native enzyme.


Subject(s)
Calpain/genetics , Amino Acid Sequence , Animals , Baculoviridae/genetics , Base Sequence , Calcium/metabolism , Calpain/isolation & purification , Calpain/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , Enzyme Activation , Erythrocytes/enzymology , Humans , Hydrolysis , Molecular Sequence Data , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spodoptera
12.
Bioorg Med Chem ; 6(5): 509-22, 1998 May.
Article in English | MEDLINE | ID: mdl-9629465

ABSTRACT

Calpain I, an intracellular cysteine protease, has been implicated in the neurodegeneration following an episode of cerebral ischemia. In this paper, we report on a series of peptidomimetic ketomethylene and carbamethylene inhibitors of recombinant human calpain I (rh calpain I). Our study reveals that the -NHCO-moiety (possible hydrogen-bonding site) at the P2-P3 region of a potent tripeptide or a dipeptide inhibitor of calpain I is not a strict requirement for enzyme recognition. Compounds 7d ((R)-2-isobutyl-4-oxo-4-(9-xanthenyl)butanoic acid ((S)-1-formyl-3-methyl)butyl amide), 31 ((R)-2-isobutyl-4-(2-sulfonylnaphthyl)butyric acid ((S)1-formyl-3-methyl)butyl amide) and 34 ((R)-2-isobutyl-4-(2-sulfoxylnaphthyl)butyric acid ((S)-1-formyl-3-methyl)butyl amide) which exhibited good activity in the enzyme assay, also inhibited calpain I in a human cell line.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Dipeptides/chemistry , Oligopeptides/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Humans , Magnetic Resonance Spectroscopy , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Protein Conformation , Recombinant Proteins/antagonists & inhibitors , Tumor Cells, Cultured
13.
J Neurochem ; 73(5): 1901-12, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537048

ABSTRACT

The c-Jun N-terminal kinase signaling cascade appears to play a role in some cases of cell death, including neuronal apoptosis. CEP-1347 (KT7515), an indolocarbazole of the K252a family, blocks this stress signaling cascade and promotes survival. Here, we used CEP-1347 to probe whether neuronal death pathways activated by distinct insults also possess elements in common. Cultured rat sympathetic neurons and neuronally differentiated PC12 cells were induced to die by withdrawal of nerve growth factor, exposure to ultraviolet irradiation, or subjection to oxidative stress. In each case, death was prevented by 100-200 nM CEP-1347. Moreover, in each of these death paradigms, c-Jun N-terminal kinase 1 activity in neuronally differentiated PC12 cells was elevated by two- or threefold, and this increase was totally blocked by CEP-1347 at concentrations that promoted survival. In contrast, 200 nM CEP-1347 did not block death due to serum withdrawal from undifferentiated PC12 cells or to activation of Fas in Jurkat T cell cultures, even though in each case c-Jun N-terminal kinase 1 activation occurred and was inhibited by CEP-1347. These observations suggest that some but not all death pathways triggered by different insults can include a common mechanistic component, a likely candidate for which is activation of the c-Jun N-terminal kinase signaling cascade.


Subject(s)
Carbazoles/pharmacology , Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Ganglia, Sympathetic/cytology , Indoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neurons/physiology , Animals , Cell Differentiation , Cells, Cultured , Enzyme Activation/drug effects , Humans , Jurkat Cells , MAP Kinase Kinase 4 , Neurites/physiology , Oxidative Stress , PC12 Cells , Rats , Rats, Sprague-Dawley , Signal Transduction , fas Receptor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL