Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuroimage ; 132: 1-7, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26876472

RESUMO

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.


Assuntos
Alcoolismo/metabolismo , Azidas/farmacocinética , Benzodiazepinas/farmacocinética , Encéfalo/metabolismo , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores de GABA-A/metabolismo , Adulto , Marcadores de Afinidade/farmacocinética , Radioisótopos de Carbono , Hipocampo/metabolismo , Humanos , Masculino , Memória , Núcleo Accumbens/metabolismo , Tomografia por Emissão de Pósitrons
2.
Neuropharmacology ; 100: 66-75, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26220312

RESUMO

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'.


Assuntos
Espinhas Dendríticas/fisiologia , Endocitose , Zíper de Leucina , Microscopia de Fluorescência/métodos , Receptores de AMPA/fisiologia , Coloração e Rotulagem/métodos , Marcadores de Afinidade/farmacocinética , Animais , Células Cultivadas , Corantes Fluorescentes/farmacocinética , Hipocampo/citologia , Hipocampo/fisiologia , Camundongos , Estrutura Terciária de Proteína , Transporte Proteico , Prótons
3.
Spectrochim Acta A Mol Biomol Spectrosc ; 143: 309-18, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25766241

RESUMO

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.


Assuntos
Marcadores de Afinidade/química , Antineoplásicos/química , Receptores ErbB/antagonistas & inibidores , Quinazolinonas/química , Marcadores de Afinidade/farmacocinética , Marcadores de Afinidade/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Receptores ErbB/metabolismo , Células MCF-7 , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Quinazolinonas/farmacocinética , Quinazolinonas/farmacologia , Coelhos , Distribuição Tecidual
4.
J Control Release ; 186: 32-40, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-24815420

RESUMO

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.


Assuntos
Marcadores de Afinidade/administração & dosagem , Carbocianinas/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Sondas Moleculares/administração & dosagem , Neoplasias/metabolismo , Receptor de Endotelina A/metabolismo , Albumina Sérica/metabolismo , Marcadores de Afinidade/química , Marcadores de Afinidade/farmacocinética , Animais , Disponibilidade Biológica , Carbocianinas/química , Carbocianinas/farmacocinética , Linhagem Celular Tumoral , Feminino , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Humanos , Camundongos Nus , Imagem Molecular/métodos , Sondas Moleculares/química , Sondas Moleculares/farmacocinética
5.
Mol Cell Endocrinol ; 337(1-2): 96-100, 2011 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-21315799

RESUMO

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.


Assuntos
Marcadores de Afinidade/farmacologia , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Recombinantes/metabolismo , Tri-Iodotironina/análogos & derivados , Marcadores de Afinidade/farmacocinética , Animais , Células COS , Chlorocebus aethiops , Humanos , Iodeto Peroxidase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Ratos , Simportadores , Tri-Iodotironina/farmacocinética , Tri-Iodotironina/farmacologia
6.
Brain Res ; 1165: 15-20, 2007 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-17662260

RESUMO

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.


Assuntos
Encéfalo/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Receptores de GABA-A/metabolismo , Marcadores de Afinidade/farmacocinética , Animais , Azidas/farmacocinética , Benzodiazepinas/farmacocinética , Ligação Competitiva/efeitos dos fármacos , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Imunoprecipitação , Masculino , Muscimol/farmacocinética , Ensaio Radioligante , Cintilografia , Ratos , Ratos Sprague-Dawley , Trítio/farmacocinética
7.
Neuropharmacology ; 52(2): 395-404, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17045310

RESUMO

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.


Assuntos
Benzofuranos/farmacologia , Ligação Competitiva/efeitos dos fármacos , Imidazóis/farmacologia , Imidazolinas/metabolismo , Monoaminoxidase/metabolismo , Marcadores de Afinidade/farmacocinética , Análise de Variância , Animais , Autorradiografia/métodos , Benzofuranos/farmacocinética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Imidazóis/farmacocinética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos CBA , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Serotonina/farmacologia , Fatores de Tempo , Trítio/farmacocinética
8.
Curr Alzheimer Res ; 3(3): 259-66, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16842103

RESUMO

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.


Assuntos
Marcadores de Afinidade/metabolismo , Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/metabolismo , Corantes Fluorescentes , Tiazóis , Marcadores de Afinidade/síntese química , Marcadores de Afinidade/farmacocinética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Animais , Benzotiazóis , Ligação Competitiva , Radioisótopos de Carbono/química , Radioisótopos de Carbono/farmacocinética , Diagnóstico por Imagem/métodos , Avaliação Pré-Clínica de Medicamentos , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Humanos , Radioisótopos do Iodo/química , Radioisótopos do Iodo/farmacocinética , Camundongos , Estrutura Molecular , Emaranhados Neurofibrilares/diagnóstico por imagem , Placa Amiloide/diagnóstico por imagem , Ligação Proteica , Cintilografia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Tiazóis/síntese química , Tiazóis/química , Tiazóis/farmacocinética
9.
Neuroscience ; 139(2): 661-70, 2006 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-16476524

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Receptor A2A de Adenosina/fisiologia , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D3/fisiologia , Marcadores de Afinidade/farmacocinética , Animais , Autorradiografia/métodos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacocinética , Hibridização In Situ/métodos , Mazindol/farmacocinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/fisiologia , Proteínas de Transporte de Nucleosídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/deficiência , Receptor A2A de Adenosina/genética , Receptores de Dopamina D1/deficiência , Receptores de Dopamina D1/genética , Receptores de Dopamina D3/deficiência , Receptores de Dopamina D3/genética , Tioinosina/análogos & derivados , Tioinosina/farmacocinética , Trítio/farmacocinética
10.
Synapse ; 55(4): 270-9, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15668983

RESUMO

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.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/análise , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Encéfalo/enzimologia , Inibidores de Fosfodiesterase/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Rolipram/metabolismo , Marcadores de Afinidade/metabolismo , Marcadores de Afinidade/farmacocinética , Animais , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Radioisótopos de Carbono , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Técnicas In Vitro , Masculino , Inibidores de Fosfodiesterase/farmacocinética , Ratos , Ratos Sprague-Dawley , Rolipram/farmacocinética , Estereoisomerismo , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Sus scrofa
11.
J Affect Disord ; 75(1): 65-9, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12781352

RESUMO

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.


Assuntos
Transtorno Bipolar/metabolismo , Transtorno Bipolar/patologia , Citalopram/farmacocinética , Hipocampo/metabolismo , Hipocampo/patologia , Proteínas de Membrana Transportadoras , Proteínas do Tecido Nervoso , Receptores de Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Marcadores de Afinidade/farmacocinética , Sítios de Ligação , Biomarcadores , Proteínas de Transporte/metabolismo , Contagem de Células , Técnicas de Cultura , Humanos , Ketanserina/farmacocinética , Glicoproteínas de Membrana/metabolismo , Antagonistas da Serotonina/farmacocinética , Proteínas da Membrana Plasmática de Transporte de Serotonina , Agonistas do Receptor de Serotonina/farmacocinética , Sumatriptana/farmacocinética
12.
J Biol Chem ; 278(15): 13196-206, 2003 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-12560326

RESUMO

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.


Assuntos
Marcadores de Afinidade/farmacologia , Aziridinas/farmacologia , Ativação do Canal Iônico/fisiologia , Porinas/fisiologia , Pregnanolona/farmacologia , Telencéfalo/metabolismo , Marcadores de Afinidade/farmacocinética , Animais , Aziridinas/farmacocinética , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Feminino , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Muscimol/farmacocinética , Oócitos/fisiologia , Porinas/análise , Pregnanolona/análogos & derivados , Pregnanolona/farmacocinética , Ratos , Proteínas Recombinantes/metabolismo , Esteroides/farmacocinética , Transfecção , Canal de Ânion 1 Dependente de Voltagem , Canais de Ânion Dependentes de Voltagem , Xenopus laevis , Ácido gama-Aminobutírico/farmacologia
13.
J Biol Chem ; 276(31): 28650-8, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11356832

RESUMO

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.


Assuntos
Proteína Relacionada ao Hormônio Paratireóideo , Hormônio Paratireóideo/metabolismo , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteínas/metabolismo , Proteínas/farmacologia , Receptores de Hormônios Paratireóideos/química , Receptores de Hormônios Paratireóideos/metabolismo , Marcadores de Afinidade/farmacocinética , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Reagentes de Ligações Cruzadas , Brometo de Cianogênio , AMP Cíclico/metabolismo , Humanos , Radioisótopos do Iodo , Modelos Moleculares , Mutagênese Sítio-Dirigida , Hormônio Paratireóideo/química , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/química , Fenilalanina/análogos & derivados , Fenilalanina/farmacocinética , Estrutura Secundária de Proteína , Proteínas/química , Ensaio Radioligante , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Transfecção
14.
J Biol Chem ; 276(24): 21199-208, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11287418

RESUMO

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.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacocinética , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacocinética , Azidas/farmacocinética , Vanadatos/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/química , Marcadores de Afinidade/farmacocinética , Animais , Sítios de Ligação , Linhagem Celular , Membrana Celular/metabolismo , Células HeLa , Humanos , Hidrólise , Insetos , Cinética , Radioisótopos de Fósforo , Proteínas Recombinantes/metabolismo , Termodinâmica , Transfecção
15.
Bioorg Med Chem Lett ; 10(20): 2275-8, 2000 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-11055337

RESUMO

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.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Marcadores de Afinidade/síntese química , Benzofenonas , Benzofenonas/síntese química , Succinimidas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Marcadores de Afinidade/química , Marcadores de Afinidade/farmacocinética , Benzofenonas/química , Benzofenonas/farmacocinética , Desenho de Fármacos , Humanos , Modelos Moleculares , Conformação Molecular , Polienos/química , Polienos/farmacocinética , Trítio
16.
J Neurochem ; 75(4): 1493-501, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10987829

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Endocitose/fisiologia , Agonistas do Receptor Purinérgico P1 , Receptores Purinérgicos P1/metabolismo , Adenosina/farmacocinética , Marcadores de Afinidade/farmacocinética , Animais , Ligação Competitiva/efeitos dos fármacos , Células CHO , Membrana Celular/metabolismo , Concanavalina A/farmacologia , Cricetinae , Relação Dose-Resposta a Droga , Endocitose/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Soluções Hipertônicas/farmacologia , Imuno-Histoquímica , Radioisótopos do Iodo/análise , Ligantes , Potássio/metabolismo , Ensaio Radioligante , Receptor A3 de Adenosina , Receptores Purinérgicos P1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Temperatura , Transfecção
17.
Mol Cell Endocrinol ; 156(1-2): 179-88, 1999 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-10612436

RESUMO

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.


Assuntos
Marcadores de Afinidade/farmacocinética , Hormônio Liberador de Gonadotropina/análogos & derivados , Receptores LHRH/metabolismo , Marcadores de Afinidade/síntese química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Ligação Competitiva , Células COS , Linhagem Celular , Reagentes de Ligações Cruzadas , Hormônio Liberador de Gonadotropina/síntese química , Hormônio Liberador de Gonadotropina/farmacocinética , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Fosfatos de Inositol/metabolismo , Cinética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Secundária de Proteína , Ensaio Radioligante , Receptores LHRH/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ovinos , Transfecção
18.
J Biol Chem ; 273(51): 34594-602, 1998 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-9852131

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Ciclo Celular/fisiologia , Marcadores de Afinidade/farmacocinética , Substituição de Aminoácidos , Anticorpos Monoclonais , Azidas/farmacocinética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/química , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacocinética , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Substâncias Macromoleculares , Mitose , Mutagênese Sítio-Dirigida , Radioisótopos de Fósforo , Fosforilação , Reação em Cadeia da Polimerase , Proteínas Recombinantes/análise , Proteínas Recombinantes/metabolismo , Frações Subcelulares/enzimologia , Transfecção
19.
J Biol Chem ; 273(48): 31916-23, 1998 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-9822661

RESUMO

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.


Assuntos
Benzoquinonas/metabolismo , Escherichia coli/enzimologia , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Estrutura Secundária de Proteína , Succinato Desidrogenase/química , Succinato Desidrogenase/metabolismo , Marcadores de Afinidade/farmacocinética , Sequência de Aminoácidos , Substituição de Aminoácidos , Azidas/farmacocinética , Sequência de Bases , Sítios de Ligação , Mapeamento Cromossômico , Cromossomos Bacterianos , Complexo II de Transporte de Elétrons , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multienzimáticos/genética , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos , Óperon , Oxirredutases/genética , Fragmentos de Peptídeos/química , Mutação Puntual , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Succinato Desidrogenase/genética , Ubiquinona/análogos & derivados , Ubiquinona/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA