Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
ACS Pharmacol Transl Sci ; 7(7): 1937-1950, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-39022353

RESUMO

Hypoparathyroidism is a common sequela of thyroid surgery; in this study, we aimed at exploring the pathogenesis behind it. The following premises suggest that wound fluid might be a causative agent. (i) Parathyroid hormone secretion is under feedback control by the calcium-sensing receptor, which responds to a diverse array of activating ligands. (ii) Postoperative hypoparathyroidism arises from a secretory deficiency of the parathyroid glands. Even in patients later unaffected by hypoparathyroidism, parathyroid hormone levels drop within hours after surgery. (iii) Wound fluid is bound to enter the tissue around the thyroid bed, where the parathyroid glands are located. Its composition is shaped by a series of proteolytic reactions triggered by wounding. Using thyroid drainage as a surrogate, we addressed the possibility that wound fluid contains compounds activating the calcium-sensing receptor. Drainage fluid ultrafiltrate was found to be rich in amino acids, and on separation by HPLC, compounds activating the calcium-sensing receptor partitioned with hydrophilic matter that rendered buffer acidic. The data show that glutamate and aspartate at millimolar concentrations supported activation of the calcium-sensing receptor, an effect contingent on low pH. In the presence of glutamate/aspartate, protons activated the calcium-sensing receptor with a pH50 of 6.1, and at pH 5, produced maximal activation. This synergistic mode of action was exclusive; glutamine/asparagine did not substitute for the acidic amino acids, nor did Ca2+ substitute for protons. NPS-2143, a negative allosteric receptor modulator completely blocked receptor activation by glutamate/aspartate and by fractionated drainage fluid. Thus, wound fluid may be involved in suppressing parathyroid hormone secretion.

2.
Mol Pharmacol ; 98(3): 250-266, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32817461

RESUMO

In medium-size, spiny striatal neurons of the direct pathway, dopamine D1- and adenosine A1-receptors are coexpressed and are mutually antagonistic. Recently, a mutation in the gene encoding the A1-receptor (A1R), A1R-G279S7.44, was identified in an Iranian family: two affected offspring suffered from early-onset l-DOPA-responsive Parkinson's disease. The link between the mutation and the phenotype is unclear. Here, we explored the functional consequence of the G279S substitution on the activity of the A1-receptor after heterologous expression in HEK293 cells. The mutation did not affect surface expression and ligand binding but changed the susceptibility to heat denaturation: the thermodynamic stability of A1R-G279S7.44 was enhanced by about 2 and 8 K when compared with wild-type A1-receptor and A1R-Y288A7.53 (a folding-deficient variant used as a reference), respectively. In contrast, the kinetic stability was reduced, indicating a lower energy barrier for conformational transitions in A1R-G279S7.44 (73 ± 23 kJ/mol) than in wild-type A1R (135 ± 4 kJ/mol) or in A1R-Y288A7.53 (184 ± 24 kJ/mol). Consistent with this lower energy barrier, A1R-G279S7.44 was more effective in promoting guanine nucleotide exchange than wild-type A1R. We detected similar levels of complexes formed between D1-receptors and wild-type A1R or A1R-G279S7.44 by coimmunoprecipitation and bioluminescence resonance energy transfer. However, lower concentrations of agonist were required for half-maximum inhibition of dopamine-induced cAMP accumulation in cells coexpressing D1-receptor and A1R-G279S7.44 than in those coexpressing wild-type A1R. These observations predict enhanced inhibition of dopaminergic signaling by A1R-G279S7.44 in vivo, consistent with a pathogenic role in Parkinson's disease. SIGNIFICANCE STATEMENT: Parkinson's disease is caused by a loss of dopaminergic input from the substantia nigra to the caudate nucleus and the putamen. Activation of the adenosine A1-receptor antagonizes responses elicited by dopamine D1-receptor. We show that this activity is more pronounced in a mutant version of the A1-receptor (A1R-G279S7.44), which was identified in individuals suffering from early-onset Parkinson's disease.


Assuntos
Substituição de Aminoácidos , Doença de Parkinson/genética , Receptor A1 de Adenosina/química , Receptor A1 de Adenosina/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Mutação , Ligação Proteica , Conformação Proteica , Estabilidade Proteica , Receptor A1 de Adenosina/genética , Termodinâmica
4.
Int J Mol Sci ; 18(11)2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29135937

RESUMO

Many diseases arise from mutations, which impair protein folding. The study of folding-deficient variants of G protein-coupled receptors and solute carrier 6 (SLC6) transporters has shed light on the folding trajectory, how it is monitored and how misfolding can be remedied. Reducing the temperature lowers the energy barrier between folding intermediates and thereby eliminates stalling along the folding trajectory. For obvious reasons, cooling down is not a therapeutic option. One approach to rescue misfolded variants is to use membrane-permeable orthosteric ligands. Antagonists of GPCRs are-in many instances-effective pharmacochaperones: they restore cell surface expression provided that they enter cells and bind to folding intermediates. Pharmacochaperoning of SLC6 transporters is less readily achieved because the ionic conditions in the endoplasmic reticulum (ER) are not conducive to binding of typical inhibitors. The second approach is to target the heat-shock protein (HSP) relay, which monitors the folding trajectory on the cytosolic side. Importantly, orthosteric ligands and HSP-inhibitors are not mutually exclusive. In fact, pharmacochaperones and HSP-inhibitors can act in an additive or synergistic manner. This was exemplified by rescuing disease-causing, folding-deficient variants of the human dopamine transporters with the HSP70 inhibitor pifithrin-µ and the pharmacochaperone noribogaine in Drosophila melanogaster.


Assuntos
Proteínas Mutantes/metabolismo , Dobramento de Proteína , Receptores Acoplados a Proteínas G/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Animais , Humanos , Modelos Biológicos
5.
Cancer Chemother Pharmacol ; 80(4): 673-684, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28779264

RESUMO

PURPOSE: Small-molecule inhibitors of heat-shock protein 90 (HSP90) have been under development as chemotherapeutic agents. The adverse events reported from early clinical trials included hyponatremia. Given the limited number of patients enrolled, the number of hyponatremia incidents was remarkable and repeatedly, the event was judged as severe. Inappropriate V2 vasopressin receptor stimulation is an established cause of hyponatremia. We explored the hypothesis that HSP90 inhibition produces hypersensitivity to vasopressin by upregulating V2-receptors. METHODS: Experiments were carried out in cell culture using HEK293 cells with heterologous expression of the human V2-receptor and HELA cells with an endogenous V2-receptor complement. We tested the effect of HSP90 inhibition by three structurally unrelated compounds (alvespimycin, luminespib, radicicol) and asserted its specificity in cells depleted of cytosolic HSP90 (by RNA interference). Assays encompassed surface V2-receptor density and vasopressin-stimulated formation of cyclic AMP (cAMP). RESULTS: The results demonstrate a twofold increase in cell-surface receptor density following pre-incubation with each of the HSP90 inhibitors. The effect had a concentration-dependence consistent with the individual potencies to inhibit HSP90. Similarly, depletion of cytosolic HSP90 increased surface-receptor density and at the same time, reduced the inhibitor effect. Upregulated V2-receptors were fully functional; hence, in culture treated with an HSP90 inhibitor, addition of vasopressin resulted in higher levels of cAMP than in controls. CONCLUSION: Since formation of cAMP is the first signalling step in raising water permeability of the collecting duct epithelia, we suggest that V2-receptor upregulation generates hypersensitivity to vasopressin linking HSP90 inhibition to the development of hyponatremia.


Assuntos
AMP Cíclico/metabolismo , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Hiponatremia/etiologia , Receptores de Vasopressinas/genética , Vasopressinas/metabolismo , Benzoquinonas/farmacologia , Citosol/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Células HeLa , Humanos , Isoxazóis/farmacologia , Lactamas Macrocíclicas/farmacologia , Macrolídeos/farmacologia , Interferência de RNA , Resorcinóis/farmacologia , Regulação para Cima
6.
Eur J Nucl Med Mol Imaging ; 44(10): 1732-1741, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28555333

RESUMO

PURPOSE: This prospective study was to investigate the value of [11C]-acetate PET and [18F]-FDG PET in the evaluation of hepatocellular carcinoma (HCC) before and after treatment with transarterial chemoembolization (TACE) and vascular endothelial growth factor (VEGF) antibody (bevacizumab). METHODS: Twenty-two patients (three women, 19 men; 62 ± 8 years) with HCC verified by histopathology were treated with TACE and bevacizumab (n = 11) or placebo (n = 11). [11C]-acetate PET and [18F]-FDG PET were performed before and after TACE with bevacizumab or placebo. Comparisons between groups were performed with t-tests and Chi-squared tests, where appropriate. Overall survival (OS) was defined as the time from start of bevacizumab or placebo until the date of death/last follow-up, respectively. RESULTS: The patient-related sensitivity of [11C]-acetate PET, [18F]-FDG PET, and combined [11C]-acetate and [18F]-FDG PET was 68%, 45%, and 73%, respectively. There was a significantly higher rate of conversion from [11C]-acetate positive lesions to negative lesions in patients treated with TACE and bevacizumab as compared with that in patients with TACE and placebo (p < 0.05). In patients with negative acetate PET, the mean OS in patients treated with TACE and bevacizumab was 259 ± 118 days and was markedly shorter as compared with that (668 ± 217 days) in patients treated with TACE and placebo (p < 0.05). In patients treated with TACE and placebo, there was significant difference in mean OS in patients with positive FDG PET as compared with that in patients with negative FDG PET (p < 0.05). The HCC lesions had different tracer avidities showing the heterogeneity of HCC. CONCLUSIONS: Our study suggests that combining [18F]-FDG with [11C]-acetate PET could be useful for the management of HCC patients and might also provide relevant prognostic and molecular heterogeneity information.


Assuntos
Acetatos , Bevacizumab/uso terapêutico , Carbono , Carcinoma Hepatocelular/terapia , Quimioembolização Terapêutica , Fluordesoxiglucose F18 , Neoplasias Hepáticas/terapia , Tomografia por Emissão de Pósitrons , Idoso , Artérias , Bevacizumab/administração & dosagem , Carcinoma Hepatocelular/diagnóstico por imagem , Feminino , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Análise de Sobrevida , Resultado do Tratamento
7.
J Pharmacol Exp Ther ; 359(1): 73-81, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27451411

RESUMO

Blockage of ß1-adrenergic receptors is one of the most effective treatments in cardiovascular medicine. Esmolol was introduced some three decades ago as a short-acting ß1-selective antagonist. Landiolol is a more recent addition. Here we compared the two compounds for their selectivity for ß1-adrenergic receptors over ß2-adrenergic receptors, partial agonistic activity, signaling bias, and pharmacochaperoning action by using human embryonic kidney (HEK)293 cell lines, which heterologously express each human receptor subtype. The affinity of landiolol for ß1-adrenergic receptors and ß2-adrenergic receptors was higher and lower than that of esmolol, respectively, resulting in an improved selectivity (216-fold versus 30-fold). The principal metabolite of landiolol (M1) was also ß1-selective, but its affinity was very low. Both landiolol and esmolol caused a very modest rise in cAMP levels but a robust increase in the phosphorylation of extracellular signal regulated kinases 1 and 2, indicating that the two drugs exerted partial agonist activity with a signaling bias. If cells were incubated for ≥24 hours in the presence of ≥1 µM esmolol, the levels of ß1-adrenergic-but not of ß2-adrenergic-receptors increased. This effect was contingent on export of the ß1-receptor from endoplasmic reticulum and was not seen in the presence of landiolol. On the basis of these observations, we conclude that landiolol offers the advantage of: 1) improved selectivity and 2) the absence of pharmacochaperoning activity, which sensitizes cells to rebound effects upon drug discontinuation.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Agonismo Parcial de Drogas , Morfolinas/farmacologia , Propanolaminas/farmacologia , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Ureia/análogos & derivados , Pressão Sanguínea/efeitos dos fármacos , AMP Cíclico/metabolismo , Células HEK293 , Frequência Cardíaca/efeitos dos fármacos , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Propranolol/farmacologia , Ureia/farmacologia
8.
Mol Pharmacol ; 87(1): 39-51, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25354767

RESUMO

Cell-permeable orthosteric ligands can assist folding of G protein-coupled receptors in the endoplasmic reticulum (ER); this pharmacochaperoning translates into increased cell surface levels of receptors. Here we used a folding-defective mutant of human A1-adenosine receptor as a sensor to explore whether endogenously produced adenosine can exert a chaperoning effect. This A1-receptor-Y(288)A was retained in the ER of stably transfected human embryonic kidney 293 cells but rapidly reached the plasma membrane in cells incubated with an A1 antagonist. This was phenocopied by raising intracellular adenosine levels with a combination of inhibitors of adenosine kinase, adenosine deaminase, and the equilibrative nucleoside transporter: mature receptors with complex glycosylation accumulated at the cell surface and bound to an A1-selective antagonist with an affinity indistinguishable from the wild-type A1 receptor. The effect of the inhibitor combination was specific, because it did not result in enhanced surface levels of two folding-defective human V2-vasopressin receptor mutants, which were susceptible to pharmacochaperoning by their cognate antagonist. Raising cellular adenosine levels by subjecting cells to hypoxia (5% O2) reproduced chaperoning by the inhibitor combination and enhanced surface expression of A1-receptor-Y(288)A within 1 hour. These findings were recapitulated for the wild-type A1 receptor. Taken together, our observations document that endogenously formed adenosine can chaperone its cognate A1 receptor. This results in a positive feedback loop that has implications for the retaliatory metabolite concept of adenosine action: if chaperoning by intracellular adenosine results in elevated cell surface levels of A1 receptors, these cells will be more susceptible to extracellular adenosine and thus more likely to cope with metabolic distress.


Assuntos
Adenosina/metabolismo , Retículo Endoplasmático/metabolismo , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Antagonistas do Receptor A1 de Adenosina/metabolismo , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Chaperonas Moleculares/metabolismo , Mutação , Plasmócitos/metabolismo , Dobramento de Proteína , Estresse Fisiológico/efeitos dos fármacos
9.
J Biol Chem ; 289(13): 9263-74, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24509856

RESUMO

The A2A receptor is a class A/rhodopsin-like G protein-coupled receptor. Coupling to its cognate protein, Gs, occurs via restricted collision coupling and is contingent on the presence of cholesterol. Agonist activation slows diffusion of the A2A adenosine receptor in the lipid bilayer. We explored the contribution of the hydrophobic core and of the extended C terminus by examining diffusion of quantum dot-labeled receptor variants in dissociated hippocampal neurons. Single particle tracking of the A2A receptor(1-311), which lacks the last 101 residues, revealed that agonist-induced confinement was abolished and that the agonist-induced decrease in diffusivity was reduced substantially. A fragment comprising the SH3 domain and the guanylate kinase domain of synapse-associated protein 102 (SAP102) was identified as a candidate interactor that bound to the A2A receptor C terminus. Complex formation between the A2A receptor and SAP102 was verified by coimmunoprecipitation and by tracking its impact on receptor diffusion. An analysis of all trajectories by a hidden Markov model was consistent with two diffusion states where agonist activation reduced the transition between the two states and, thus, promoted the accumulation of the A2A receptor in the compartment with slow mobility. Overexpression of SAP102 precluded the access of the A2A receptor to a compartment with restricted mobility. In contrast, a mutated A2A receptor (with (383)DVELL(387) replaced by RVRAA) was insensitive to the action of SAP102. These observations show that the hydrophobic core per se does not fully account for the agonist-promoted change in mobility of the A2A receptor. The extended carboxyl terminus allows for regulatory input by scaffolding molecules such as SAP102.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Hipocampo/citologia , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Receptor A2A de Adenosina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Difusão/efeitos dos fármacos , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Cadeias de Markov , Mutação , Ratos , Receptor A2A de Adenosina/química , Receptor A2A de Adenosina/genética
10.
J Biol Chem ; 288(40): 28831-44, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-23965991

RESUMO

The adenosine A2A receptor is a prototypical rhodopsin-like G protein-coupled receptor but has several unique structural features, in particular a long C terminus (of >120 residues) devoid of a palmitoylation site. It is known to interact with several accessory proteins other than those canonically involved in signaling. However, it is evident that many more proteins must interact with the A2A receptor, if the trafficking trajectory of the receptor is taken into account from its site of synthesis in the endoplasmic reticulum (ER) to its disposal by the lysosome. Affinity-tagged versions of the A2A receptor were expressed in HEK293 cells to identify interacting partners residing in the ER by a proteomics approach based on tandem affinity purification. The receptor-protein complexes were purified in quantities sufficient for analysis by mass spectrometry. We identified molecular chaperones (heat-shock proteins HSP90α and HSP70-1A) that interact with and retain partially folded A2A receptor prior to ER exit. Complex formation between the A2A receptor and HSP90α (but not HSP90ß) and HSP70-1A was confirmed by co-affinity precipitation. HSP90 inhibitors also enhanced surface expression of the receptor in PC12 cells, which endogenously express the A2A receptor. Finally, proteins of the HSP relay machinery (e.g. HOP/HSC70-HSP90 organizing protein and P23/HSP90 co-chaperone) were recovered in complexes with the A2A receptor. These observations are consistent with the proposed chaperone/coat protein complex II exchange model. This posits that cytosolic HSP proteins are sequentially recruited to folding intermediates of the A2A receptor. Release of HSP90 is required prior to recruitment of coat protein complex II components. This prevents premature ER export of partially folded receptors.


Assuntos
Citoplasma/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Receptor A2A de Adenosina/metabolismo , Transdução de Sinais , Animais , Citoplasma/efeitos dos fármacos , Células HEK293 , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Humanos , Imunoprecipitação , Oxirredutases Intramoleculares/metabolismo , Espectrometria de Massas , Células PC12 , Prostaglandina-E Sintases , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Ligação Proteica/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Ubiquitina-Proteína Ligases/metabolismo
11.
Subcell Biochem ; 63: 1-21, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23161130

RESUMO

The polypeptide of a G protein-coupled receptor is inserted into the membrane of the endoplasmic reticulum while being translated and this process by itself may be sufficient to establish the proper receptor fold. X-ray structures reveal a common polypeptide topology with little variation in the alignment and orientation of the seven transmembrane segments, the proximal carboxyl terminus (C-tail) and parts of the extracellular loops. These define a structural core the stability of which probably represents a major criterion for the receptor to pass endoplasmic reticulum (ER) quality control; point mutations affecting the structure of the core have an extraordinary chance of causing receptor retention. In contrast, cytoplasmic loops 2 and 3 and the distal C-tail are poorly ordered at least in the absence of an interaction partner. Similarly, the amino terminal tail of rhodopsin-related receptors (but not of receptor subtypes where ligand binding requires a stable fold of the N-tail) is unlikely to establish a stable fold. These segments can cause ER retention when mutated to inappropriately expose hydrophobic peptide patches; to prevent protein aggregation chaperone molecules attach to them thus initiating selection for ER-associated degradation. It is less clear however if there are additional mechanisms to specifically survey the transmembrane core at the level of the lipid bilayer or if insufficient packing is detected due to misalignment of the cytoplasmic or extracellular face of the receptor.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
12.
Purinergic Signal ; 8(4): 677-92, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22476939

RESUMO

Neuroblastoma SH-SY5Y (SH) cells endogenously express A(2A) adenosine receptors and can be differentiated into a sympathetic neuronal phenotype, capable of depolarisation-dependent noradrenaline release. Using differentiated SH culture, we here explored the link between A(2A)-receptor signalling and neurotransmitter release. In response to the receptor agonist CGS21680, the cells produced cyclic AMP (cAMP), and when depolarised, they released increased amounts of noradrenaline. An A(2A)-receptor antagonist, XAC, as well as an inhibitor of cAMP-dependent protein kinase A (PKA), H89, depressed agonist-dependent release. In the presence of XAC or H89, noradrenaline release was found to be below basal values. This suggested that release facilitation also owes to constitutive receptor activity. We demonstrate that even in the absence of an agonist, the native A(2A)-receptor stimulated cAMP production, leading to the activation of PKA and enhanced noradrenaline release. Ancillary, non-cAMP-dependent effects of the receptor (i.e. phosphorylation of CREB, of Rabphilin3A) were refractory to constitutive activation. PKA-dependent facilitation of noradrenaline release was recapitulated with membrane-permeable 8-Br-cAMP; in addition to facilitation, 8-Br-cAMP caused marked inhibition of release, an effect not observed upon receptor activation. Inhibition by receptor-independent cAMP was likely due to suppression of voltage-dependent calcium current (VDCC) and increased activity of Src-family kinases. Receptor-mediated release facilitation was reproduced in the presence of tetrodotoxin (blocking action potentials); hence, the signalling occurred at the active zone comprising release sites. Our findings thus support (1) presynaptic localisation of the A(2A)-receptor and (2) suggest that compartmentalised pathways transmit cAMP signalling in order to facilitate depolarisation-dependent neurotransmitter release.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Norepinefrina/metabolismo , Receptor A2A de Adenosina/metabolismo , Transdução de Sinais , Adenosina/análogos & derivados , Adenosina/farmacologia , Diferenciação Celular/fisiologia , Linhagem Celular , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fenetilaminas/farmacologia , Transmissão Sináptica/efeitos dos fármacos
13.
Neuropharmacology ; 62(4): 1708-16, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22155208

RESUMO

Amantadine is an established antiparkinsonian drug with a still unclear molecular site of action. In vivo studies on rodents, in vitro studies on tissue of rodents as well as binding studies on post mortem human tissue implicate monoamine transporters and NMDA receptors. In order to re-examine its action at human variants of these proteins on intact cells we established cells stably expressing the human NR1/2A NMDA-receptor, noradrenaline transporter (NAT) or dopamine transporter (DAT) and tested the activity of amantadine in patch-clamp, uptake, release, and cytotoxicity experiments. Amantadine was less potent in blockade of NMDA-induced inward currents than in blockade of noradrenaline uptake and in induction of inward currents in NAT expressing cells. It was 30 times more potent in blocking uptake in NAT- than in DAT cells. Amantadine induced NAT-mediated release at concentrations of 10-100 µM in superfusion experiments and blocked NAT-mediated cytotoxicity of the parkinsonism inducing neurotoxin 1-methyl-4-phenyl-pyridinium (MPP(+)) at concentrations of 30-300 µM, whereas 300-1000 µM amantadine was necessary to block NMDA-receptor mediated cytotoxicity. Similar to amphetamine, amantadine was inactive at α(2A)-adrenergic receptors and induced reverse noradrenaline transport by NAT albeit with smaller effect size. Thus, amantadine acted as "amphetamine-like releaser" with selectivity for the noradrenergic system. These findings and differences with memantine, which had been reported as less efficient antiparkinsonian drug than amantadine but in our hands was significantly more potent at the NMDA-receptor, suggest contributions from a noradrenergic mechanism in the antiparkinsonian action of amantadine.


Assuntos
Amantadina/farmacologia , Antiparkinsonianos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Linhagem Celular , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Células HEK293 , Humanos , N-Metilaspartato/farmacologia , Receptores Adrenérgicos alfa 2/metabolismo
14.
Mol Pharmacol ; 77(6): 940-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20219842

RESUMO

Exchanging each of the conserved aromatic residues of the NPxxY(x)(5,6)F sequence (at the boundary of helices 7 and 8) generated variants of the A(1) adenosine receptor that were retained within the cell. The mutations disconnected a link between alpha-helix 7 and cytosolic helix 8, likely destabilizing the structure of the proximal carboxyl terminus. The mutant receptors were rescued by incubation of cells with a pharmacochaperone, a membrane-permeable ligand that homosterically binds to the receptor; pharmacochaperoning restored the density of functional receptors at the plasma membrane. The following observations support the assumption that retention and the site of pharmacochaperone action were within bounds of the endoplasmic reticulum (ER): 1) the retained receptor colocalized with an ER marker; 2) pharmacochaperoning initiated receptor transfer to Golgi stacks; and 3) the inhibitor of glycoprotein synthesis tunicamycin suppressed receptor chaperoning. Our data are consistent with the hypothesis that pharmacochaperoning stabilizes the structure of late folding intermediates and lifts a block on maturation, allowing the receptors to exit from the ER. We suggest that the ER-associated 40-kDa heat shock protein family member D(1) receptor interacting protein 78 (DRiP78; M(r), approximately 78,000) represents a model executor of quality control. Overexpressed DRiP78 interacted physically with the A(1) receptor, inhibited export to the plasma membrane, and in this action was selective for the mutants relative to the wild-type receptor. Both agonist and antagonist were effective chaperone ligands. Thus, occupancy of the binding pocket corrected the mutation-induced disorder, indicating a mutual impingement of the transmembrane domain and the proximal carboxyl terminus in establishing the stable receptor fold.


Assuntos
Retículo Endoplasmático/efeitos dos fármacos , Chaperonas Moleculares/metabolismo , Receptor A1 de Adenosina/metabolismo , Animais , Linhagem Celular , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Complexo de Golgi/metabolismo , Humanos , Ligantes , Mutagênese , Mutação , Ratos , Receptor A1 de Adenosina/genética , Tunicamicina/farmacologia
15.
Breast Care (Basel) ; 3(6): 401-406, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-21048911

RESUMO

Adjuvant therapy improves survival in breast cancer patients. However, both antihormonal agents and cytostatic chemotherapy meet with variable success. We have searched the literature for biological causes of variability in drug response. Evidence suggests that additional markers may be introduced because of their potentially predictive value in adjuvant therapy: i) overexpression of epidermal growth factor receptor is likely inversely correlated to the sensitivity to estrogen antagonists; ii) presence of the GAB2 adaptor protein and of the ABCC3 and mdr-1 efflux pumps modulates taxane sensitivity in HER2-positive breast cancer; and iii) CYP2D6 genotyping should be a routine measure to avoid failure of tamox-ifen treatment. In contrast, there is little in the way of genetic evidence for differences in the pharmacokinetics of other antihormonal or cytostatic drugs. Nevertheless, genotypes may affect efficacy and toxicity of cytostatic drugs (e.g. doxorubicin), but this evidence has to be confirmed by prospective trials.

16.
Mol Pharmacol ; 69(1): 397-405, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16210429

RESUMO

The receptor-mimetic peptide D2N, derived from the cytoplasmic domain of the D(2) dopamine receptor, activates G protein alpha-subunits (G(i) and G(o)) directly. Using D2N, we tested the current hypotheses on the mechanism of receptor-mediated G protein activation, which differ by the role assigned to the Gbetagamma-subunit: 1) a receptor-prompted movement of Gbetagamma is needed to open up the nucleotide exit pathway ("gear-shift" and "lever-arm" model) or 2) the receptor first engages Gbetagamma and then triggers GDP release by interacting with the carboxyl (C) terminus of Galpha (the "sequential-fit" model). Our results with D2N were compatible with the latter hypothesis. D2N bound to the extreme C terminus of the alpha-subunit and caused a conformational change that was transmitted to the switch regions. Hence, D2N led to a decline in the intrinsic tryptophan fluorescence, increased the guanine nucleotide exchange rate, and modulated the Mg(2+) control of nucleotide binding. A structural alteration in the outer portion of helix alpha5 (substitution of an isoleucine by proline) blunted the stimulatory action of D2N. This confirms that helix alpha5 links the guanine nucleotide binding pocket to the receptor contact site on the G protein. However, neither the alpha-subunit amino terminus (as a lever-arm) nor Gbetagamma was required for D2N-mediated activation; conversely, assembly of the Galphabetagamma heterotrimer stabilized the GDP-bound species and required an increased D2N concentration for activation. We propose that the receptor can engage the C terminus of the alpha-subunit to destabilize nucleotide binding from the "back side" of the nucleotide binding pocket.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/química , Magnésio/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Espectrometria de Fluorescência
17.
Mol Pharmacol ; 69(4): 1083-94, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16339847

RESUMO

Many membrane proteins incur a folding problem during biosynthesis; only a fraction thereof is exported from the endoplasmic reticulum (ER), because quality control is stringent. This is also true for G protein-coupled receptors. Here, we identify the deubiquitinating enzyme Usp4 as an interaction partner of the A2a adenosine receptor, a Gs-coupled receptor. Usp4 binds to the carboxyl terminus of the A2A receptor and allows for its accumulation as deubiquinated protein. This relaxes ER quality control and enhances cell surface expression of functionally active receptor. The effect of Usp4 on the A2A receptor was specific because 1) it was not seen in C-terminally truncated versions of the receptor; 2) it was not mimicked by Usp14, another member of the ubiquitin-specific protease family; and 3) it was not seen with the metabotropic glutamate receptor-5, another G protein-coupled receptor with a high propensity for intracellular retention. These observations show that deubiquinating enzymes can regulate quality control in the ER.


Assuntos
Proteínas Oncogênicas/metabolismo , Receptores A2 de Adenosina/metabolismo , Ubiquitina/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Primers do DNA , Retículo Endoplasmático/metabolismo , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Ensaio Radioligante , Ratos , Ubiquitina Tiolesterase , Proteases Específicas de Ubiquitina
18.
Methods Mol Biol ; 259: 183-95, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15250493

RESUMO

The activation of heterotrimeric G-proteins is tightly regulated by the exchange of GTP for GDP in the alpha-subunit; mostly--but not exclusively--seven-transmembrane receptors function as the guanine nucleotide exchange factors (GEFs). A research goal may be to determine which G-protein alpha-subunit is activated by the receptor under investigation. In a membrane preparation obtained from cells or tissues this can be achieved in a seemingly straightforward manner by determining if the receptor increases the covalent incorporation of GTP analogs into G-protein alpha-subunits. Because the GTP analogs may be labeled to high specific radioactivity the alpha-subunit can then be identified with the use of specific antibodies. One of the compounds we present here (2',3'-dialdehyde-GTP) can also be employed to block receptor-mediated G-protein activation and to disrupt the cognate signaling pathway.


Assuntos
Marcadores de Afinidade/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/análogos & derivados , Animais , Azidas/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Oxirredução , Radioisótopos de Fósforo/metabolismo
19.
Neuropsychopharmacology ; 28(7): 1317-27, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12784121

RESUMO

The adenosine A(2A) receptor and the dopamine D(2) receptor are prototypically coupled to G(s) and G(i)/G(o), respectively. In striatal intermediate spiny neurons, these receptors are colocalized in dendritic spines and act as mutual antagonists. This antagonism has been proposed to occur at the level of the receptors or of receptor-G protein coupling. We tested this model in PC12 cells which endogenously express A(2A) receptors. The human D(2) receptor was introduced into PC12 cells by stable transfection. A(2A)-agonist-mediated inhibition of D(2) agonist binding was absent in PC12 cell membranes but present in HEK293 cells transfected as a control. However, in the resulting PC12 cell lines, the action of the D(2) agonist quinpirole depended on the expression level of the D(2) receptor: at low and high receptor levels, the A(2A)-agonist-induced elevation of cAMP was enhanced and inhibited, respectively. Forskolin-stimulated cAMP formation was invariably inhibited by quinpirole. The effects of quinpirole were abolished by pretreatment with pertussis toxin. A(2A)-receptor-mediated cAMP formation was inhibited by other G(i)/G(o)-coupled receptors that were either endogenously present (P(2y12)-like receptor for ADP) or stably expressed after transfection (A(1) adenosine, metabotropic glutamate receptor-7A). Similarly, voltage activated Ca(2+) channels were inhibited by the endogenous P(2Y) receptor and by the heterologously expressed A(1) receptor but not by the D(2) receptor. These data indicate functional segregation of signaling components. Our observations are thus compatible with the proposed model that D(2) and A(2A) receptors are closely associated, but they highlight the fact that this interaction can also support synergism.


Assuntos
Difosfato de Adenosina/análogos & derivados , Adenosina/análogos & derivados , Adenilil Ciclases/metabolismo , Receptores de Dopamina D2/fisiologia , Receptores Purinérgicos P1/fisiologia , Proteínas de Xenopus , 4-(3-Butoxi-4-metoxibenzil)-2-imidazolidinona/farmacologia , Adenina/farmacocinética , Adenosina/farmacocinética , Adenosina/farmacologia , Adenosina Desaminase/farmacologia , Difosfato de Adenosina/farmacologia , Animais , Benzamidas/farmacocinética , Ligação Competitiva , Ciclo Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cofilina 1 , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas do Citoesqueleto , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacocinética , Antagonistas dos Receptores de Dopamina D2 , Ativação Enzimática , Expressão Gênica , Ácido Glutâmico/farmacologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Células PC12 , Técnicas de Patch-Clamp/métodos , Toxina Pertussis/farmacologia , Fenetilaminas/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Fosfoproteínas/farmacologia , Propionatos/farmacologia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Pirrolidinas/farmacocinética , Quimpirol/farmacologia , Ensaio Radioligante/métodos , Ratos , Receptores de Dopamina D2/genética , Receptores Purinérgicos P1/genética , Tionucleotídeos/farmacologia , Transfecção , Triazinas/farmacocinética , Triazóis/farmacocinética
20.
J Biol Chem ; 278(32): 30283-93, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12764156

RESUMO

The carboxyl terminus (C-tail) of G protein-coupled receptors is divergent in length and structure and may represent an individualized cytoplasmic domain. By progressively truncating the A1 adenosine receptor, a Gi/o-coupled receptor with short cytoplasmic stretches, we identify two inherent functions of the C-tail, namely a role in receptor export from the endoplasmic reticulum (ER) and a role in G protein coupling. Deletion of the last 22 and 26 amino acids (of 36) reduced and completely abolished surface expression of the receptor, respectively. The severely truncated receptors were retained in the ER and failed to bind ligands. If overexpressed, even a substantial portion of the full-length receptor was retained in the ER in a form that was not functional. These data indicate that folding is rate limiting in export from the ER and that the proximal segment of the carboxyl terminus provides a docking site for the machinery involved in folding and quality control. In addition, the proximal portion is also important in G protein coupling. This latter role was unmasked when the distal portion of the C-tail (the extreme 18 amino acids, including a palmitoylated cysteine) had been removed; the resulting receptor was functional and transferred the agonist-mediated signal more efficiently than the full-length receptor. Signaling was enhanced because the coupling affinity increased (by 3-fold), which translated into a higher agonist potency. Thus, the distal portion of the carboxyl terminus provides for an autoinhibitory restraint, presumably by folding back and preventing G protein access to the proximal part of the C-tail.


Assuntos
Receptores Purinérgicos P1/química , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Citoplasma/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Ligantes , Sistema de Sinalização das MAP Quinases , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Testes de Precipitina , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA