Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Neuroimage ; 132: 1-7, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26876472

ABSTRACT

The importance of the GABA-benzodiazepine receptor complex and its subtypes are increasingly recognised in addiction. Using the α1/α5 benzodiazepine receptor PET radioligand [(11)C]Ro15 4513, we previously showed reduced binding in the nucleus accumbens and hippocampus in abstinent alcohol dependence. We proposed that reduced [(11)C]Ro15 4513 binding in the nucleus accumbens was a marker of addiction whilst the reduction in hippocampus and positive relationship with memory was a consequence of chronic alcohol abuse. To examine this further we assessed [(11)C]Ro15 4513 binding in another addiction, opiate dependence, and used spectral analysis to estimate contributions of α1 and α5 subtypes to [(11)C]Ro15 4513 binding in opiate and previously acquired alcohol-dependent groups. Opiate substitute maintained opiate-dependent men (n=12) underwent an [(11)C]Ro15 4513 PET scan and compared with matched healthy controls (n=13). We found a significant reduction in [(11)C]Ro15 4513 binding in the nucleus accumbens in the opiate-dependent compared with the healthy control group. There was no relationship between [(11)C]Ro15 4513 binding in the hippocampus with memory. We found that reduced [(11)C]Ro15 4513 binding was associated with reduced α5 but not α1 subtypes in the opiate-dependent group. This was also seen in an alcohol-dependent group where an association between memory performance and [(11)C]Ro15 4513 binding was primarily driven by α5 and not α1 subtype. We suggest that reduced α5 levels in the nucleus accumbens are associated with addiction since we have now shown this in dependence to two pharmacologically different substances, alcohol and opiates.


Subject(s)
Alcoholism/metabolism , Azides/pharmacokinetics , Benzodiazepines/pharmacokinetics , Brain/metabolism , Opioid-Related Disorders/metabolism , Receptors, GABA-A/metabolism , Adult , Affinity Labels/pharmacokinetics , Carbon Radioisotopes , Hippocampus/metabolism , Humans , Male , Memory , Nucleus Accumbens/metabolism , Positron-Emission Tomography
2.
Neuropharmacology ; 100: 66-75, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26220312

ABSTRACT

Techniques to visualize receptor trafficking in living neurons are important, but currently available methods are limited in their labeling efficiency, specificity and reliability. Here we report a method for receptor labeling with a basic leucine zipper domain peptide (ZIP) and a binding cassette specific to ZIP. Receptors are tagged with a ZIP-binding cassette at their extracellular domain. Tagged receptors expressed in cultured cells were labeled with exogenously applied fluorescently labeled ZIP with low background and high affinity. To test if ZIP labeling is useful in monitoring endocytosis and intracellular trafficking, we next conjugated ZIP with a pH-sensitive dye RhP-M (ZIP-RhP-M). ZIP binding to its binding cassette was pH-resistant and RhP-M fluorescence dramatically increased in acidic environment. Thus AMPA-type glutamate receptors (AMPARs) labeled by ZIP-RhP-M can report receptor endocytosis and subsequent intracellular trafficking. Application of ZIP-RhP-M to cultured hippocampal neurons expressing AMPARs tagged with a ZIP-binding cassette resulted in appearance of fluorescent puncta in PSD-95-positive large spines, suggesting local endocytosis and acidification of AMPARs in individual mature spines. This spine pool of AMPARs in acidic environment was distinct from the early endosomes labeled by transferrin uptake. These results suggest that receptor labeling by ZIP-RhP-M is a useful technique for monitoring endocytosis and intracellular trafficking. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.


Subject(s)
Dendritic Spines/physiology , Endocytosis , Leucine Zippers , Microscopy, Fluorescence/methods , Receptors, AMPA/physiology , Staining and Labeling/methods , Affinity Labels/pharmacokinetics , Animals , Cells, Cultured , Fluorescent Dyes/pharmacokinetics , Hippocampus/cytology , Hippocampus/physiology , Mice , Protein Structure, Tertiary , Protein Transport , Protons
3.
Spectrochim Acta A Mol Biomol Spectrosc ; 143: 309-18, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25766241

ABSTRACT

The binding capabilities of a series of novel quinazolinone molecules were established and stated in a comprehensive computational methodology as well as by in vitro analysis. The main focus of this work was to achieve more insight of the interactions with crystal structure of PDB ID: 1M17 and predict their binding mode to EGFR. Three molecules were screened for further examination, which were synthesized and characterized using spectroscopic techniques. The persuasive affinity of these molecules towards EGFR inhibition (IC50 for QT=45nM) was established and validated from specific kinase assay including the cell viability spectrophotometric assay (QT=12nM). Drug likeliness property were also considered by analysing, the ADME of these molecules by using scintigraphic techniques. The result showed antitumour activity of QT (4.17 tumour/muscle at 4h). Further photo physical properties were also analysed to see in vitro HSA binding to QT.


Subject(s)
Affinity Labels/chemistry , Antineoplastic Agents/chemistry , ErbB Receptors/antagonists & inhibitors , Quinazolinones/chemistry , Affinity Labels/pharmacokinetics , Affinity Labels/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , ErbB Receptors/metabolism , MCF-7 Cells , Mice, Inbred BALB C , Molecular Docking Simulation , Quinazolinones/pharmacokinetics , Quinazolinones/pharmacology , Rabbits , Tissue Distribution
4.
J Control Release ; 186: 32-40, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24815420

ABSTRACT

Small molecular imaging probes are often found to be rapidly cleared from the circulation. In order to improve signal to noise ratio (SNR) by high probe accumulation in the target tissue we intended to prolong the presence of the probes in the circulation by exploiting inherent transport mechanisms. Human serum albumin (HSA) is playing an increasingly important role as a drug carrier in clinical settings and drugs directly bound to albumin or attached to albumin binding moieties have been successfully developed for treatment approaches. To optimize the bioavailability of existing fluorescent probes, a hydrophobic affinity tag is installed, which enhances albumin binding. In a first experiment an endothelin-A receptor (ETAR) probe is modified by inserting a trivalent linker, attaching an albumin affinity tag and labeling the conjugate with the fluorescent dye Cy 5.5. The spectroscopic properties of the conjugate are examined by photometer- and fluorometer measurements in comparison to a probe without albumin binding tag. Albumin binding was proven by agarose gel electrophoresis. The affinity towards ETAR was confirmed in vitro by cell binding assays on human fibrosarcoma cells (HT-1080) and in vivo by murine xenograft imaging studies. In vitro, the modified probe retains high target binding in the absence and presence of albumin. Binding could be blocked by predosing with ETAR antagonist atrasentan, proving specificity. The in vivo examinations in comparison to the established probe showed a reduced renal elimination and a prolonged circulation of the tracer resulting in significantly higher signal intensity (SI) at the target and a higher signal-to-noise ratio (SNR) between 3h and 96 h after injection. In summary, we designed a small molecular, non-peptidic fluorescent probe which targets ETAR and reversibly binds to serum albumins. The reversible binding to albumin enhances the biological half-life of the probe substantially and enables near infrared optical imaging of subcutaneous tumors for several days. This approach of reversibly attaching probes to serum albumin may serve as a tool to optimize tracer distribution for more precise target characterization in molecular imaging experiments.


Subject(s)
Affinity Labels/administration & dosage , Carbocyanines/administration & dosage , Fluorescent Dyes/administration & dosage , Molecular Probes/administration & dosage , Neoplasms/metabolism , Receptor, Endothelin A/metabolism , Serum Albumin/metabolism , Affinity Labels/chemistry , Affinity Labels/pharmacokinetics , Animals , Biological Availability , Carbocyanines/chemistry , Carbocyanines/pharmacokinetics , Cell Line, Tumor , Female , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Humans , Mice, Nude , Molecular Imaging/methods , Molecular Probes/chemistry , Molecular Probes/pharmacokinetics
5.
Mol Cell Endocrinol ; 337(1-2): 96-100, 2011 Apr 30.
Article in English | MEDLINE | ID: mdl-21315799

ABSTRACT

Thyroid hormone (TH) transporter proteins mediate transport of TH across the plasma membrane, thereby facilitating its intracellular bioavailability. As only a few transporters have been identified which are relatively specific for TH, including monocarboxylate transporter (MCT) 8 and MCT10, the need for identification of novel specific TH transporters is obvious. A possible strategy to identify TH transporters is their modification with a ligand-derived affinity-label and subsequent identification by mass spectrometry. Previously, N-bromoacetyl (BrAc)-iodothyronines have been reported as useful affinity-labels for human (h) MCT8. In the present study we reinvestigated possible BrAc[(125)I]T3-labeling of hMCT8 and hMCT10. The present study demonstrates that hMCT8 and hMCT10 both facilitate BrAc[(125)I]T3 transport, but are not labeled by BrAc[(125)I]T3. We provide evidence that human protein disulfide isomerase, which molecular mass is similar to hMCT8, is labeled by BrAc[(125)I]T3. In addition, differential inhibitory effects were observed of iodothyronines derivatives with different side chains on T3 transport by hMCT8 and hMCT10. In conclusion, we demonstrated that not hMCT8 and hMCT10, but human protein disulfide isomerase, is labeled by BrAc[(125)I]T3. The usefulness of BrAc[(125)I]T3 as a tool for the identification of novel TH transporters remains to be explored.


Subject(s)
Affinity Labels/pharmacology , Amino Acid Transport Systems, Neutral/metabolism , Monocarboxylic Acid Transporters/metabolism , Recombinant Proteins/metabolism , Triiodothyronine/analogs & derivatives , Affinity Labels/pharmacokinetics , Animals , COS Cells , Chlorocebus aethiops , Humans , Iodide Peroxidase/metabolism , Protein Disulfide-Isomerases/metabolism , Rats , Symporters , Triiodothyronine/pharmacokinetics , Triiodothyronine/pharmacology
6.
Brain Res ; 1165: 15-20, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17662260

ABSTRACT

In the present study, we investigated the co-localization pattern of the delta subunit with other subunits of GABA(A) receptors in the rat brain using immunoprecipitation and Western blotting techniques. Furthermore, we investigated whether low concentrations of ethanol affect the delta-subunit-containing GABA(A) receptor assemblies in the rat brain using radioligand binding to the rat brain membrane homogenates as well as to the immunoprecipitated receptor assemblies. Our results revealed that delta subunit is not co-localized with gamma(2) subunit but it is associated with the alpha(1), alpha(4) or alpha(6), beta(2) and/or beta(3) subunit(s) of GABA(A) receptors in the rat brain. Ethanol (1-50 mM) neither affected [(3)H]muscimol (3 nM) binding nor diazepam-insensitive [(3)H]Ro 15-4513 (2 nM) binding in the rat cerebellum and cerebral cortex membranes. However, a higher concentration of ethanol (500 mM) inhibited the binding of these radioligands to the GABA(A) receptors partially in the rat cerebellum and cerebral cortex. Similarly, ethanol (up to 50 mM) did not affect [(3)H]muscimol (15 nM) binding to the immunoprecipitated delta-subunit-containing GABA(A) receptor assemblies in the rat cerebellum and hippocampus but it inhibited the binding partially at a higher concentration (500 mM). These results suggest that the native delta-subunit-containing GABA(A) receptors do not play a major role in the pharmacology of clinically relevant low concentrations of ethanol.


Subject(s)
Brain/drug effects , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Receptors, GABA-A/metabolism , Affinity Labels/pharmacokinetics , Animals , Azides/pharmacokinetics , Benzodiazepines/pharmacokinetics , Binding, Competitive/drug effects , Brain/diagnostic imaging , Brain/metabolism , Dose-Response Relationship, Drug , Immunoprecipitation , Male , Muscimol/pharmacokinetics , Radioligand Assay , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Tritium/pharmacokinetics
7.
Neuropharmacology ; 52(2): 395-404, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17045310

ABSTRACT

BU99006 is an irreversible I(2) ligand which selectively inactivates I(2) binding sites, making it an ideal tool with which to study I(2) site mechanism. We sought to determine the effects of BU99006 on I(2) binding in relation to monoamine oxidase (MAO), and the time course of these effects. In vitro, rat brain membranes that were pre-treated with 10 microM BU99006 showed no change in MAO activity, despite suffering a significant reduction in [(3)H]2BFI binding (52.5+/-19.6 to 8.5+/-3.8 fmol mg(-1), 84%). Furthermore, reversible I(2) ligands 2BFI and BU224 were able to inhibit MAO, whether treated with BU99006 or not. In vivo, a 5 mg kg(-1) i.v. dose of BU99006 in rats rapidly reduced [(3)H]2BFI binding with similar magnitude (85%, maximal reduction after 20 min), without effect on either MAO activity or the specific binding of selective MAO-A and MAO-B radioligands. Moreover, following this irreversible treatment, recovery of central [(3)H]2BFI binding occurred with a rapid half-life of 4.3 h in rat brain (2.0 h in mouse), which is not consistent with a site on MAO. These data indicate that the high affinity site which is occupied by [(3)H]2BFI and irreversibly binds BU99006, is not the same as that which causes inhibition of MAO, and may point to the existence of another I(2) binding site.


Subject(s)
Benzofurans/pharmacology , Binding, Competitive/drug effects , Imidazoles/pharmacology , Imidazolines/metabolism , Monoamine Oxidase/metabolism , Affinity Labels/pharmacokinetics , Analysis of Variance , Animals , Autoradiography/methods , Benzofurans/pharmacokinetics , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Female , Imidazoles/pharmacokinetics , In Vitro Techniques , Male , Mice , Mice, Inbred CBA , Rats , Rats, Wistar , Reaction Time/drug effects , Serotonin/pharmacology , Time Factors , Tritium/pharmacokinetics
8.
Curr Alzheimer Res ; 3(3): 259-66, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16842103

ABSTRACT

Lipophilic analogs of thioflavin S were synthesized and radiolabeled with positron or single photon emitting radionuclides. The binding affinity for Abeta was evaluated using isolated amyloid fibrils from human brain tissue. Binding specificity was assessed using fluorescent tissue staining. In vivo brain uptake was evaluated in mice. Following synthesis, neutral analogs of thioflavin S capable of radiolabeling with (11)C or (125)I, were found to bind isolated human Abeta with affinities in the nanomolar range. Fluorescent tissue staining showed selective binding to Abeta deposits in vitro. Biodistribution of selected compounds displayed high brain permeability at early time points. At later points, the compounds were cleared from the normal brain, indicating low non-specific binding in vivo. These studies indicated that novel amyloid imaging probes can be developed based on thioflavin S that readily entered the brain and selectively bound to Abeta deposits and neurofibrilary tangles. Potential applications of these amyloid binding agents include facilitating drug screening in animal models and use as in vivo markers of early and definitive diagnosis of AD.


Subject(s)
Affinity Labels/metabolism , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Fluorescent Dyes , Thiazoles , Affinity Labels/chemical synthesis , Affinity Labels/pharmacokinetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Animals , Benzothiazoles , Binding, Competitive , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacokinetics , Diagnostic Imaging/methods , Drug Evaluation, Preclinical , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Humans , Iodine Radioisotopes/chemistry , Iodine Radioisotopes/pharmacokinetics , Mice , Molecular Structure , Neurofibrillary Tangles/diagnostic imaging , Plaque, Amyloid/diagnostic imaging , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Thiazoles/chemical synthesis , Thiazoles/chemistry , Thiazoles/pharmacokinetics
9.
Neuroscience ; 139(2): 661-70, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16476524

ABSTRACT

Dopamine and adenosine receptors are known to share a considerable overlap in their regional distribution, being especially rich in the basal ganglia. Dopamine and adenosine receptors have been demonstrated to exhibit a parallel distribution on certain neuronal populations, and even when not directly co-localized, relationships (both antagonistic and synergistic) have been described. This study was designed to investigate dopaminergic and purinergic systems in mice with ablations of individual dopamine or adenosine receptors. In situ hybridization histochemistry and autoradiography was used to examine the level of mRNA and protein expression of specific receptors and transporters in dopaminergic pathways. Expression of the mRNA encoding the dopamine D2 receptor was elevated in the caudate putamen of D1, D3 and A2A receptor knockout mice; this was mirrored by an increase in D2 receptor protein in D1 and D3 receptor knockout mice, but not in A2A knockout mice. Dopamine D1 receptor binding was decreased in the caudate putamen, nucleus accumbens, olfactory tubercle and ventral pallidum of D2 receptor knockout mice. In substantia nigra pars compacta, dopamine transporter mRNA expression was dramatically decreased in D3 receptor knockout mice, but elevated in A2A receptor knockout mice. All dopamine receptor knockout mice examined exhibited increased A2A receptor binding in the caudate putamen, nucleus accumbens and olfactory tubercle. These data are consistent with the existence of functional interactions between dopaminergic and purinergic systems in these reward and motor-related brain regions.


Subject(s)
Brain/metabolism , Receptor, Adenosine A2A/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D3/physiology , Affinity Labels/pharmacokinetics , Animals , Autoradiography/methods , Brain/anatomy & histology , Brain/drug effects , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacokinetics , In Situ Hybridization/methods , Mazindol/pharmacokinetics , Mice , Mice, Inbred C57BL , Mice, Knockout/physiology , Nucleoside Transport Proteins/metabolism , Protein Binding/drug effects , RNA, Messenger/metabolism , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Receptors, Dopamine D1/deficiency , Receptors, Dopamine D1/genetics , Receptors, Dopamine D3/deficiency , Receptors, Dopamine D3/genetics , Thioinosine/analogs & derivatives , Thioinosine/pharmacokinetics , Tritium/pharmacokinetics
10.
Synapse ; 55(4): 270-9, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15668983

ABSTRACT

Cyclic AMP (cAMP) is a continually produced nucleotide which is inactivated by hydrolysis to 5'AMP via phosphodiesterase 4 (PDE4) enzymes. Rolipram is a selective PDE4 inhibitor which exists in two enantiomeric forms, R(-) and S(+). Both of these enantiomers have previously been labelled with carbon-11 and used as positron emission tomography (PET) ligands for measuring PDE4 expression and function, and indirectly to explore the function of the cAMP second messenger, in vivo, using PET. The aim of these studies was to relate the in vitro affinities of the two rolipram enantiomers using standard pharmacological assays with the in vivo behaviour of the two enantiomers using PET. In vitro competition assays were performed using rat cortical membranes and [(3)H]R(-)- and [(3)H]S(+)-rolipram with increasing concentrations of either unlabelled R(-)- or S(+)-rolipram. In vivo, a series of PET studies were performed in the porcine brain using [(11)C]R(-)-rolipram with co-administration of increasing doses of either unlabelled R(-)- or S(+)-rolipram. Additional in vivo PET studies were performed using [(11)C]S(+)-rolipram with saturating doses of rolipram. In all studies, R(-)-rolipram exhibited a higher affinity for the PDE4 enzyme than S(+)-rolipram. The calculated affinity ratios were 7.97 from the in vitro studies; 12.5 from the in vivo studies using [(11)C]R(-)-rolipram; and 14.7 from the in vivo studies using [(11)C]S(+)-rolipram. To conclude, the in vitro affinities of R(-)- and S(+)-rolipram predict their apparent in vivo behaviour in the porcine brain, as measured by PET.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/analysis , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Brain/enzymology , Phosphodiesterase Inhibitors/metabolism , Positron-Emission Tomography/methods , Rolipram/metabolism , Affinity Labels/metabolism , Affinity Labels/pharmacokinetics , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/anatomy & histology , Brain/drug effects , Carbon Radioisotopes , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , In Vitro Techniques , Male , Phosphodiesterase Inhibitors/pharmacokinetics , Rats , Rats, Sprague-Dawley , Rolipram/pharmacokinetics , Stereoisomerism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Sus scrofa
11.
J Affect Disord ; 75(1): 65-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12781352

ABSTRACT

BACKGROUND: Various studies suggest the hippocampus and serotonergic systems are important in the pathology of bipolar disorder (BD). We therefore measured hippocampal serotonergic markers in post-mortem tissue from BD and control subjects. METHODS: The density and affinity of [3H]citalopram binding to the serotonin transporter (SERT), as well as the density of the 5HT(2A), 5HT(1A), 5HT(1D) and 5HT(1F) receptors were measured. RESULTS: The density of SERT and 5HT receptors was no different in BD. There was a significant decrease in the affinity of [3H]citalopram binding to SERT in the stratum lacunosum-moleculare (S(lac)) in BD (K(d) mean+/-S.E.M.=4.3+/-0.8 vs. 1.9+/-0.3 nM). LIMITATIONS: This study was completed using relatively small cohorts. CONCLUSIONS: There are no generalised changes in hippocampal serotonergic markers in the hippocampus from subjects with BD. There is a decreased affinity of radioligand binding to S(lac) SERT in subjects with BD.


Subject(s)
Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Citalopram/pharmacokinetics , Hippocampus/metabolism , Hippocampus/pathology , Membrane Transport Proteins , Nerve Tissue Proteins , Receptors, Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Affinity Labels/pharmacokinetics , Binding Sites , Biomarkers , Carrier Proteins/metabolism , Cell Count , Culture Techniques , Humans , Ketanserin/pharmacokinetics , Membrane Glycoproteins/metabolism , Serotonin Antagonists/pharmacokinetics , Serotonin Plasma Membrane Transport Proteins , Serotonin Receptor Agonists/pharmacokinetics , Sumatriptan/pharmacokinetics
12.
J Biol Chem ; 278(15): 13196-206, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12560326

ABSTRACT

Neuroactive steroids modulate the function of gamma-aminobutyric acid, type A (GABA(A)) receptors in the central nervous system by an unknown mechanism. In this study we have used a novel neuroactive steroid analogue, 3 alpha,5 beta-6-azi-3-hydroxypregnan-20-one (6-AziP), as a photoaffinity labeling reagent to identify neuroactive steroid binding sites in rat brain. 6-AziP is an effective modulator of GABA(A) receptors as evidenced by its ability to inhibit binding of [(35)S]t-butylbicyclophosphorothionate to rat brain membranes and to potentiate GABA-elicited currents in Xenopus oocytes and human endothelial kidney 293 cells expressing GABA(A) receptor subunits (alpha(1)beta(2)gamma(2)). [(3)H]6-AziP produced time- and concentration-dependent photolabeling of protein bands of approximately 35 and 60 kDa in rat brain membranes. The 35-kDa band was half-maximally labeled at a [(3)H]6-AziP concentration of 1.9 microM, whereas the 60-kDa band was labeled at higher concentrations. The photolabeled 35-kDa protein was isolated from rat brain by two-dimensional PAGE and identified as voltage-dependent anion channel-1 (VDAC-1) by both matrix-assisted laser desorption ionization time-of-flight and ESI-tandem mass spectrometry. Monoclonal antibody directed against the N terminus of VDAC-1 immunoprecipitated labeled 35-kDa protein from a lysate of rat brain membranes, confirming that VDAC-1 is the species labeled by [(3)H]6-AziP. The beta(2) and beta(3) subunits of the GABA(A) receptor were co-immunoprecipitated by the VDAC-1 antibody suggesting a physical association between VDAC-1 and GABA(A) receptors in rat brain membranes. These data suggest that neuroactive steroid effects on the GABA(A) receptor may be mediated by binding to an accessory protein, VDAC-1.


Subject(s)
Affinity Labels/pharmacology , Aziridines/pharmacology , Ion Channel Gating/physiology , Porins/physiology , Pregnanolone/pharmacology , Telencephalon/metabolism , Affinity Labels/pharmacokinetics , Animals , Aziridines/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cell Line , Cell Membrane/drug effects , Cell Membrane/physiology , Female , Humans , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscimol/pharmacokinetics , Oocytes/physiology , Porins/analysis , Pregnanolone/analogs & derivatives , Pregnanolone/pharmacokinetics , Rats , Recombinant Proteins/metabolism , Steroids/pharmacokinetics , Transfection , Voltage-Dependent Anion Channel 1 , Voltage-Dependent Anion Channels , Xenopus laevis , gamma-Aminobutyric Acid/pharmacology
13.
J Biol Chem ; 276(31): 28650-8, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11356832

ABSTRACT

The carboxyl-terminal portions of parathyroid hormone (PTH)-(1--34) and PTH-related peptide (PTHrP)-(1-36) are critical for high affinity binding to the PTH/PTHrP receptor (P1R), but the mechanism of receptor interaction for this domain is largely unknown. To identify interaction sites between the carboxyl-terminal region of PTHrP-(1--36) and the P1R, we prepared analogs of [I(5),W(23),Y(36)]PTHrP-(1--36)-amide with individual p-benzoyl-l-phenylalanine (Bpa) substitutions at positions 22--35. When tested with LLC-PK(1) cells stably transfected with human P1R (hP1R), the apparent binding affinity and the EC(50) of agonist-stimulated cAMP accumulation for each analog was, with the exception of the Bpa(24)-substituted analog, similar to that of the parent compound. The radiolabeled Bpa(23)-, Bpa(27)-, Bpa(28)-, and Bpa(33)-substituted compounds affinity-labeled the hP1R sufficiently well to permit subsequent mapping of the cross-linked receptor region. Each of these peptides cross-linked to the amino-terminal extracellular domain of the P1R: [I(5),Bpa(23),Y(36)]PTHrP-(1-36)-amide cross-linked to the extreme end of this domain (residues 33-63); [I(5),W(23),Bpa(27),Y(36)]PTHrP-(1--36)-amide cross-linked to residues 96--102; [I(5),W(23),Bpa(28),Y(36)]PTHrP-(1--36)- amide cross-linked to residues 64--95; and [I(5),W(23), Bpa(33),Y(36)]PTHrP-(1--36)-amide cross-linked to residues 151-172. These data thus predict that residues 23, 27, 28, and 33 of native PTHrP are each near to different regions of the amino-terminal extracellular receptor domain of the P1R. This information helps define sites of proximity between several ligand residues and this large receptor domain, which so far has been largely excluded from models of the hormone-receptor complex.


Subject(s)
Parathyroid Hormone-Related Protein , Parathyroid Hormone/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Proteins/metabolism , Proteins/pharmacology , Receptors, Parathyroid Hormone/chemistry , Receptors, Parathyroid Hormone/metabolism , Affinity Labels/pharmacokinetics , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Cross-Linking Reagents , Cyanogen Bromide , Cyclic AMP/metabolism , Humans , Iodine Radioisotopes , Models, Molecular , Mutagenesis, Site-Directed , Parathyroid Hormone/chemistry , Parathyroid Hormone/pharmacology , Peptide Fragments/chemistry , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacokinetics , Protein Structure, Secondary , Proteins/chemistry , Radioligand Assay , Receptor, Parathyroid Hormone, Type 1 , Receptors, Parathyroid Hormone/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transfection
14.
J Biol Chem ; 276(24): 21199-208, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11287418

ABSTRACT

P-glycoprotein (Pgp) is an ATP-dependent drug efflux pump whose overexpression confers multidrug resistance to cancer cells. Pgp exhibits a robust drug substrate-stimulable ATPase activity, and vanadate (Vi) blocks this activity effectively by trapping Pgp nucleotide in a non-covalent stable transition state conformation. In this study we compare Vi-induced [alpha-(32)P]8-azido-ADP trapping into Pgp in the presence of [alpha-(32)P]8-azido-ATP (with ATP hydrolysis) or [alpha-(32)P]8-azido-ADP (without ATP hydrolysis). Vi mimics P(i) to trap the nucleotide tenaciously in the Pgp.[alpha-(32)P]8-azido-ADP.Vi conformation in either condition. Thus, by using [alpha-(32)P]8-azido-ADP we show that the Vi-induced transition state of Pgp can be generated even in the absence of ATP hydrolysis. Furthermore, half-maximal trapping of nucleotide into Pgp in the presence of Vi occurs at similar concentrations of [alpha-(32)P]8-azido-ATP or [alpha-(32)P]8-azido-ADP. The trapped [alpha-(32)P]8-azido-ADP is almost equally distributed between the N- and the C-terminal ATP sites of Pgp in both conditions. Additionally, point mutations in the Walker B domain of either the N- (D555N) or C (D1200N)-terminal ATP sites that arrest ATP hydrolysis and Vi-induced trapping also show abrogation of [alpha-(32)P]8-azido-ADP trapping into Pgp in the absence of hydrolysis. These data suggest that both ATP sites are dependent on each other for function and that each site exhibits similar affinity for 8-azido-ATP (ATP) or 8-azido-ADP (ADP). Similarly, Pgp in the transition state conformation generated with either ADP or ATP exhibits drastically reduced affinity for the binding of analogues of drug substrate ([(125)I]iodoarylazidoprazosin) as well as nucleotide (2'(3')-O-(2,4,6-trinitrophenyl)adenosine 5'-triphosphate). Analyses of Arrhenius plots show that trapping of Pgp with [alpha-(32)P]8-azido-ADP (in the absence of hydrolysis) displays an approximately 2.5-fold higher energy of activation (152 kJ/mol) compared with that observed when the transition state intermediate is generated through hydrolysis of [alpha-(32)P]8-azido-ATP (62 kJ/mol). In aggregate, these results demonstrate that the Pgp.[alpha-(32)P]8-azido-ADP (or ADP).Vi transition state complexes generated either in the absence of or accompanying [alpha-(32)P]8-azido-ATP hydrolysis are functionally indistinguishable.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacokinetics , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacokinetics , Azides/pharmacokinetics , Vanadates/pharmacology , ATP Binding Cassette Transporter, Subfamily B/chemistry , Affinity Labels/pharmacokinetics , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , HeLa Cells , Humans , Hydrolysis , Insecta , Kinetics , Phosphorus Radioisotopes , Recombinant Proteins/metabolism , Thermodynamics , Transfection
15.
Bioorg Med Chem Lett ; 10(20): 2275-8, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11055337

ABSTRACT

A benzophenone photoaffinity label 9 based on the polyene natural product (-)-stipiamide has been constructed using a diaminoethane spacer and the radioactive agent [3H]-BZDC (N-succinimidyl p-benzoyl-(2,3-3H)-dehydrocinnamate). Photoaffinity experiments show specific binding to human P-glycoprotein (Pgp) in the presence of cis-flupentixol but not with cyclosporin A.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Affinity Labels/chemical synthesis , Benzophenones , Benzophenones/chemical synthesis , Succinimides , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Affinity Labels/chemistry , Affinity Labels/pharmacokinetics , Benzophenones/chemistry , Benzophenones/pharmacokinetics , Drug Design , Humans , Models, Molecular , Molecular Conformation , Polyenes/chemistry , Polyenes/pharmacokinetics , Tritium
16.
J Neurochem ; 75(4): 1493-501, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10987829

ABSTRACT

A(3) adenosine receptors have been proposed to play an important role in the pathophysiology of cerebral ischemia with a regimen-dependent nature of the therapeutic effects probably related to receptor desensitization and down-regulation. Here we studied the agonist-induced internalization of human A(3) adenosine receptors in transfected Chinese hamster ovary cells, and then we evaluated the relationship between internalization and signal desensitization and resensitization. Binding of N(6)-(4-amino-3-[(125)I]iodobenzyl)adenosine-5'-N-methyluronamide to membranes from Chinese hamster ovary cells stably transfected with the human A(3) adenosine receptor showed a profile typical of these receptors in other cell lines (K:(D) = 1.3+/-0.08 nM; B(max) = 400+/-28 fmol/mg of proteins). The iodinated agonist, bound at 4 degrees C to whole transfected cells, was internalized by increasing the temperature to 37 degrees C with a rate constant of 0.04+/-0.034 min(-1). Agonist-induced internalization of A(3) adenosine receptors was directly demonstrated by immunogold electron microscopy, which revealed the localization of these receptors in plasma membranes and intracellular vesicles. Moreover, short-term exposure of these cells to the agonist caused rapid desensitization as tested in adenylyl cyclase assays. Subsequent removal of the agonist led to restoration of the receptor function and recycling of the receptors to the cell surface. The rate constant of receptor recycling was 0.02+/-0.0017 min(-1). Blockade of internalization and recycling demonstrated that internalization did not affect signal desensitization, whereas recycling of internalized receptors was implicated in the signal resensitization.


Subject(s)
Adenosine/analogs & derivatives , Endocytosis/physiology , Purinergic P1 Receptor Agonists , Receptors, Purinergic P1/metabolism , Adenosine/pharmacokinetics , Affinity Labels/pharmacokinetics , Animals , Binding, Competitive/drug effects , CHO Cells , Cell Membrane/metabolism , Concanavalin A/pharmacology , Cricetinae , Dose-Response Relationship, Drug , Endocytosis/drug effects , Humans , Hydrogen-Ion Concentration , Hypertonic Solutions/pharmacology , Immunohistochemistry , Iodine Radioisotopes/analysis , Ligands , Potassium/metabolism , Radioligand Assay , Receptor, Adenosine A3 , Receptors, Purinergic P1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Temperature , Transfection
17.
Mol Cell Endocrinol ; 156(1-2): 179-88, 1999 Oct 25.
Article in English | MEDLINE | ID: mdl-10612436

ABSTRACT

A new photoreactive gonadotropin-releasing hormone (GnRH) antagonist [Ac-(4-azidobenzoyl)-D-Lys1, D-4-Cl-Phe2, D-Trp3, D-Arg6, D-Ala10]GnRH (PAnt-1) was synthesized and shown to bind covalently to mouse and human GnRH receptors after ultraviolet irradiation. PAnt-1 exhibited high binding affinity (Ki = 3.1 +/- 0.8 nM), and high crosslinking efficiency as shown by loss of 78% of binding sites following crosslinking at saturating concentration. Crosslinking resulted in irreversible receptor blockade as shown by inhibition of GnRH-stimulated inositol phosphate production. PAnt-1 has a photoreactive group at residue 1 of the peptide, a region believed to be critical in determining antagonist versus agonist properties of GnRH analogues. The attachment site of PAnt- to the receptor was localized between residues 11 and 19 of the extracellular N-terminal domain of the receptor by peptide mapping studies using natural sequence differences between human, mouse and sheep GnRH receptors, as well as a panel of GnRH receptor constructs with a series of engineered protease cleavage sites. A disulphide bridge between Cys14 and Cys200 was cleaved during crosslinking, suggesting that Cys14 is the crosslinked residue. These results suggest that peptide GnRH antagonists bind to the receptor with the N-terminal end of the peptide positioned in a site comprising the constrained regions of the N-terminal domain and second extracellular loop in the vicinity of the Cys14-Cys200 disulphide bridge.


Subject(s)
Affinity Labels/pharmacokinetics , Gonadotropin-Releasing Hormone/analogs & derivatives , Receptors, LHRH/metabolism , Affinity Labels/chemical synthesis , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , COS Cells , Cell Line , Cross-Linking Reagents , Gonadotropin-Releasing Hormone/chemical synthesis , Gonadotropin-Releasing Hormone/pharmacokinetics , Gonadotropin-Releasing Hormone/pharmacology , Humans , Inositol Phosphates/metabolism , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Secondary , Radioligand Assay , Receptors, LHRH/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sheep , Transfection
18.
J Biol Chem ; 273(51): 34594-602, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9852131

ABSTRACT

Phosphorylation of the RII regulatory subunits of cyclic AMP-dependent protein kinases (PKAs) was examined during the HeLa cell cycle. Three RIIalpha isoforms of 51, 54, and 57 kDa were identified by RIIalpha immunodetection and labeling with 8-azido[32P]cAMP in different cell cycle phases. These isoforms were characterized as different phosphorylation states by the use of selective PKA and cyclin-directed kinase inhibitors. Whereas RIIalpha autophosphorylation by PKA caused RIIalpha to shift from 51 to 54 kDa, phosphorylation of RIIalpha by one other or a combination of several kinases activated during mitosis caused RIIalpha to shift from 51 to 57 kDa. In vivo incorporation of [32P]orthophosphate into mitotic cells and RIIalpha immunoprecipitation demonstrated that RIIalpha was hyperphosphorylated on a different site than the one phosphorylated by PKA. Deletion and mutation analysis demonstrated that the cyclin B-p34(cdc2) kinase (CDK1) phosphorylated human recombinant RIIalpha in vitro on Thr54. Whereas RIIalpha was associated with the Golgi-centrosomal region during interphase, it was dissociated from its centrosomal localization at metaphase-anaphase transition. Furthermore, particulate RIIalpha from HeLa cell extracts was solubilized following incubation with CDK1 in vitro. Our results suggest that at the onset of mitosis, CDK1 phosphorylates RIIalpha, and this may alter its subcellular localization.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Cycle/physiology , Affinity Labels/pharmacokinetics , Amino Acid Substitution , Antibodies, Monoclonal , Azides/pharmacokinetics , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacokinetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Macromolecular Substances , Mitosis , Mutagenesis, Site-Directed , Phosphorus Radioisotopes , Phosphorylation , Polymerase Chain Reaction , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Subcellular Fractions/enzymology , Transfection
19.
J Biol Chem ; 273(48): 31916-23, 1998 Nov 27.
Article in English | MEDLINE | ID: mdl-9822661

ABSTRACT

When purified ubiquinone (Q)-depleted succinate-ubiquinone reductase from Escherichia coli is photoaffinity-labeled with 3-azido-2-methyl-5-methoxy-[3H]6-geranyl-1,4-benzoquinone ([3H]azido-Q) followed by SDS-polyacrylamide gel electrophoresis, radioactivity is found in the SdhC subunit, indicating that this subunit is responsible for ubiquinone binding. An [3H]azido-Q-linked peptide, with a retention time of 61.7 min, is obtained by high performance liquid chromatography of the protease K digest of [3H]azido-Q-labeled SdhC obtained from preparative SDS-polyacrylamide gel electrophoresis on labeled reductase. The partial N-terminal amino acid sequence of this peptide is NH2-TIRFPITAIASILHRVS-, corresponding to residues 17-33. The ubiquinone-binding domain in the proposed structural model of SdhC, constructed based on the hydropathy plot of the deduced amino acid sequence of this protein, is located at the N-terminal end toward the transmembrane helix I. To identify amino acid residues responsible for ubiquinone binding, substitution mutations at the putative ubiquinone-binding region of SdhC were generated and characterized. E. coli NM256 lacking genomic succinate-Q reductase genes was constructed and used to harbor the mutated succinate-Q reductase genes in a low copy number pRKD418 plasmid. Substitution of serine 27 of SdhC with alanine, cysteine, or threonine or substitution of arginine 31 with alanine, lysine, or histidine yields cells unable to grow aerobically in minimum medium with succinate as carbon source. Furthermore, little succinate-ubiquinone reductase activity and [3H]azido-Q uptake are detected in succinate-ubiquinone reductases prepared from these mutant cells grown aerobically in LB medium. These results indicate that the hydroxyl group, the size of the amino acid side chain at position 27, and the guanidino group at position 31 of SdhC are critical for succinate-ubiquinone reductase activity, perhaps by formation of hydrogen bonds with carbonyl groups of the 1,4-benzoquinone ring of the quinone molecule. The hydroxyl group, but not the size of the amino acid side chain, at position 33 of SdhC is also important, because Ser-33 can be substituted with threonine but not with alanine.


Subject(s)
Benzoquinones/metabolism , Escherichia coli/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Structure, Secondary , Succinate Dehydrogenase/chemistry , Succinate Dehydrogenase/metabolism , Affinity Labels/pharmacokinetics , Amino Acid Sequence , Amino Acid Substitution , Azides/pharmacokinetics , Base Sequence , Binding Sites , Chromosome Mapping , Chromosomes, Bacterial , Electron Transport Complex II , Escherichia coli/genetics , Escherichia coli/growth & development , Kinetics , Models, Molecular , Molecular Sequence Data , Multienzyme Complexes/genetics , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Operon , Oxidoreductases/genetics , Peptide Fragments/chemistry , Point Mutation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Succinate Dehydrogenase/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL