Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 181(16): 2851-2868, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38657956

RESUMO

BACKGROUND AND PURPOSE: The analgesic action of paracetamol involves KV7 channels, and its metabolite N-acetyl-p-benzo quinone imine (NAPQI), a cysteine modifying reagent, was shown to increase currents through such channels in nociceptors. Modification of cysteine residues by N-ethylmaleimide, H2O2, or nitric oxide has been found to modulate currents through KV7 channels. The study aims to identify whether, and if so which, cysteine residues in neuronal KV7 channels might be responsible for the effects of NAPQI. EXPERIMENTAL APPROACH: To address this question, we used a combination of perforated patch-clamp recordings, site-directed mutagenesis, and mass spectrometry applied to recombinant KV7.1 to KV7.5 channels. KEY RESULTS: Currents through the cardiac subtype KV7.1 were reduced by NAPQI. Currents through all other subtypes were increased, either by an isolated shift of the channel voltage dependence to more negative values (KV7.3) or by such a shift combined with increased maximal current levels (KV7.2, KV7.4, KV7.5). A stretch of three cysteine residues in the S2-S3 linker region of KV7.2 was necessary and sufficient to mediate these effects. CONCLUSION AND IMPLICATION: The paracetamol metabolite N-acetyl-p-benzo quinone imine (NAPQI) modifies cysteine residues of KV7 subunits and reinforces channel gating in homomeric and heteromeric KV7.2 to KV7.5, but not in KV7.1 channels. In KV7.2, a triple cysteine motif located within the S2-S3 linker region mediates this reinforcement that can be expected to reduce the excitability of nociceptors and to mediate antinociceptive actions of paracetamol.


Assuntos
Acetaminofen , Benzoquinonas , Cisteína , Iminas , Cisteína/metabolismo , Acetaminofen/farmacologia , Benzoquinonas/farmacologia , Benzoquinonas/metabolismo , Animais , Iminas/farmacologia , Iminas/química , Iminas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Potássio KCNQ/metabolismo , Canais de Potássio KCNQ/genética , Humanos , Motivos de Aminoácidos , Analgésicos não Narcóticos/farmacologia , Células HEK293 , Ratos
2.
PLoS Negl Trop Dis ; 12(4): e0006428, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29702654

RESUMO

The parasitic liver fluke Fasciola hepatica infests mainly ruminants, but it can also cause fasciolosis in people, who ingest the metacercariae encysted on plants. The drug of choice to treat fasciolosis is triclabendazole (TBZ), which has been on the market for several decades. This is also true for the other available drugs. Accordingly, drug-resistant flukes have been emerging at an increasing rate making it desirable to identify alternative drug targets. Here, we focused on the fact that adult F. hepatica persists in the hostile environment of the bile ducts of infected organisms. A common way to render bile acids less toxic is to conjugate them to taurine (2-aminoethanesulfonic acid). We cloned a transporter from the solute carrier-6 (SLC6) family, which was most closely related to the GABA-transporter-2 of other organisms. When heterologously expressed, this F. hepatica transporter supported the high-affinity cellular uptake of taurine (KM = 12.0 ± 0.5 µM) but not of GABA. Substrate uptake was dependent on Na+- and Cl- (calculated stoichiometry 2:1). Consistent with the low chloride concentration in mammalian bile, the F. hepatica transporter had a higher apparent affinity for Cl- (EC50 = 14±3 mM) than the human taurine transporter (EC50 = 55±7 mM). We incubated flukes with unconjugated bile acids in the presence and absence of taurine: taurine promoted survival of flukes; the taurine transporter inhibitor guanidinoethansulfonic acid abolished this protective effect of taurine. Based on these observations, we conclude that the taurine transporter is critical for the survival of liver flukes in the bile. Thus, the taurine transporter represents a candidate drug target.


Assuntos
Anti-Helmínticos/farmacologia , Ácidos e Sais Biliares/farmacologia , Fasciola hepatica/genética , Fasciolíase/parasitologia , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Sequência de Aminoácidos , Animais , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Cloretos/metabolismo , Fasciola hepatica/efeitos dos fármacos , Fasciola hepatica/fisiologia , Expressão Gênica , Genes Reporter , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Filogenia , Alinhamento de Sequência , Sódio/metabolismo , Triclabendazol
3.
Nat Commun ; 8: 14089, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28102201

RESUMO

The human serotonin transporter (hSERT) mediates uptake of serotonin from the synaptic cleft and thereby terminates serotonergic signalling. We have previously found by single-molecule microscopy that SERT forms stable higher-order oligomers of differing stoichiometry at the plasma membrane of living cells. Here, we report that SERT oligomer assembly at the endoplasmic reticulum (ER) membrane follows a dynamic equilibration process, characterized by rapid exchange of subunits between different oligomers, and by a concentration dependence of the degree of oligomerization. After trafficking to the plasma membrane, however, the SERT stoichiometry is fixed. Stabilization of the oligomeric SERT complexes is mediated by the direct binding to phosphoinositide phosphatidylinositol-4,5-biphosphate (PIP2). The observed spatial decoupling of oligomer formation from the site of oligomer operation provides cells with the ability to define protein quaternary structures independent of protein density at the cell surface.


Assuntos
Fosfoinositídeo Fosfolipase C/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Animais , Células CHO , Cricetulus , Retículo Endoplasmático , Regulação da Expressão Gênica , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética
4.
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
5.
J Pharmacol Exp Ther ; 348(2): 346-58, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24307198

RESUMO

Ibogaine is a psychoactive indole alkaloid. Its use as an antiaddictive agent has been accompanied by QT prolongation and cardiac arrhythmias, which are most likely caused by human ether a go-go-related gene (hERG) potassium channel inhibition. Therefore, we studied in detail the interaction of ibogaine with hERG channels heterologously expressed in mammalian kidney tsA-201 cells. Currents through hERG channels were blocked regardless of whether ibogaine was applied via the extracellular or intracellular solution. The extent of inhibition was determined by the relative pH values. Block occurred during activation of the channels and was not observed for resting channels. With increasing depolarizations, ibogaine block grew and developed faster. Steady-state activation and inactivation of the channel were shifted to more negative potentials. Deactivation was slowed, whereas inactivation was accelerated. Mutations in the binding site reported for other hERG channel blockers (Y652A and F656A) reduced the potency of ibogaine, whereas an inactivation-deficient double mutant (G628C/S631C) was as sensitive as wild-type channels. Molecular drug docking indicated binding within the inner cavity of the channel independently of the protonation of ibogaine. Experimental current traces were fit to a kinetic model of hERG channel gating, revealing preferential binding of ibogaine to the open and inactivated state. Taken together, these findings show that ibogaine blocks hERG channels from the cytosolic side either in its charged form alone or in company with its uncharged form and alters the currents by changing the relative contribution of channel states over time.


Assuntos
Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Antagonistas de Aminoácidos Excitatórios/farmacologia , Alucinógenos/farmacologia , Ibogaína/farmacologia , Antagonistas de Entorpecentes/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Substituição de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Citosol/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Antagonistas de Aminoácidos Excitatórios/efeitos adversos , Antagonistas de Aminoácidos Excitatórios/química , Alucinógenos/efeitos adversos , Alucinógenos/química , Humanos , Concentração de Íons de Hidrogênio , Ibogaína/efeitos adversos , Ibogaína/química , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Conformação Molecular , Simulação de Acoplamento Molecular , Proteínas Mutantes/agonistas , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Antagonistas de Entorpecentes/efeitos adversos , Antagonistas de Entorpecentes/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
6.
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
7.
PLoS Comput Biol ; 9(2): e1002909, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23436987

RESUMO

The high-resolution crystal structure of the leucine transporter (LeuT) is frequently used as a template for homology models of the dopamine transporter (DAT). Although similar in structure, DAT differs considerably from LeuT in a number of ways: (i) when compared to LeuT, DAT has very long intracellular amino and carboxyl termini; (ii) LeuT and DAT share a rather low overall sequence identity (22%) and (iii) the extracellular loop 2 (EL2) of DAT is substantially longer than that of LeuT. Extracellular zinc binds to DAT and restricts the transporter's movement through the conformational cycle, thereby resulting in a decrease in substrate uptake. Residue H293 in EL2 praticipates in zinc binding and must be modelled correctly to allow for a full understanding of its effects. We exploited the high-affinity zinc binding site endogenously present in DAT to create a model of the complete transmemberane domain of DAT. The zinc binding site provided a DAT-specific molecular ruler for calibration of the model. Our DAT model places EL2 at the transporter lipid interface in the vicinity of the zinc binding site. Based on the model, D206 was predicted to represent a fourth co-ordinating residue, in addition to the three previously described zinc binding residues H193, H375 and E396. This prediction was confirmed by mutagenesis: substitution of D206 by lysine and cysteine affected the inhibitory potency of zinc and the maximum inhibition exerted by zinc, respectively. Conversely, the structural changes observed in the model allowed for rationalizing the zinc-dependent regulation of DAT: upon binding, zinc stabilizes the outward-facing state, because its first coordination shell can only be completed in this conformation. Thus, the model provides a validated solution to the long extracellular loop and may be useful to address other aspects of the transport cycle.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Zinco/metabolismo , 1-Metil-4-fenilpiridínio/química , Sequência de Aminoácidos , Sítios de Ligação , Análise Mutacional de DNA , Proteínas da Membrana Plasmática de Transporte de Dopamina/química , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência , Água/química , Zinco/química
8.
J Biol Chem ; 283(14): 9276-88, 2008 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-18218631

RESUMO

The A(2A)-adenosine receptor is a prototypical G(s) protein-coupled receptor but stimulates MAPK/ERK in a G(s)-independent way. The A(2A) receptor has long been known to undergo restricted collision coupling with G(s); the mechanistic basis for this mode of coupling has remained elusive. Here we visualized agonist-induced changes in mobility of the yellow fluorescent protein-tagged receptor by fluorescence recovery after photobleaching microscopy. Stimulation with a specific A(2A) receptor agonist did not affect receptor mobility. In contrast, stimulation with dopamine decreased the mobility of the D(2) receptor. When coexpressed in the same cell, the A(2A) receptor precluded the agonist-induced change in D(2) receptor mobility. Thus, the A(2A) receptor did not only undergo restricted collision coupling, but it also restricted the mobility of the D(2) receptor. Restricted mobility was not due to tethering to the actin cytoskeleton but was, in part, related to the cholesterol content of the membrane. Depletion of cholesterol increased receptor mobility but blunted activation of adenylyl cyclase, which was accounted for by impaired formation of the ternary complex of agonist, receptor, and G protein. These observations support the conclusion that the A(2A) receptor engages G(s) and thus signals to adenylyl cyclase in cholesterol-rich domains of the membrane. In contrast, stimulation of MAPK by the A(2A) receptor was not impaired. These findings are consistent with a model where the recruitment of these two pathways occurs in physically segregated membrane microdomains. Thus, the A(2A) receptor is the first example of a G protein-coupled receptor documented to select signaling pathways in a manner dependent on the lipid microenvironment of the membrane.


Assuntos
Membrana Celular/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Receptor A2A de Adenosina/metabolismo , Agonistas do Receptor A2 de Adenosina , Adenilil Ciclases/metabolismo , Animais , Colesterol/metabolismo , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Células PC12 , Fotodegradação , Transporte Proteico/fisiologia , Ratos , Receptores de Dopamina D2/metabolismo
9.
Nat Med ; 12(5): 568-73, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16648858

RESUMO

The growing number of recently identified negative feedback regulators of receptor tyrosine kinases (RTKs) highlights the importance of signal attenuation and modulation for correct signaling outcome. Mitogen-inducible gene 6 (Mig6 also known as RALT or Gene 33) is a multiadaptor protein thought to be involved in the regulation of RTK and stress signaling. Here, we show that deletion of the mouse gene encoding Mig6 (designated Errfi1, which stands for ERBB receptor feedback inhibitor 1) causes hyperactivation of endogenous epidermal growth factor receptor (EGFR) and sustained signaling through the mitogen-activated protein kinase (MAPK) pathway, resulting in overproliferation and impaired differentiation of epidermal keratinocytes. Furthermore, Errfi1-/- mice develop spontaneous tumors in various organs and are highly susceptible to chemically induced formation of skin tumors. A tumor-suppressive role for Mig6 is supported by our finding that MIG6 is downregulated in various human cancers. Inhibition of endogenous Egfr signaling with the Egfr inhibitor gefitinib (Iressa) or replacement of wild-type Egfr with the kinase-deficient protein encoded by the hypomorphic Egfr(wa2) allele completely rescued skin defects in Erffi1-/- mice. Carcinogen-induced tumors displayed by Errfi1-/- mice were highly sensitive to gefitinib. These results indicate that Mig6 is a specific negative regulator of Egfr signaling in skin morphogenesis and is a novel tumor suppressor of Egfr-dependent carcinogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Receptores ErbB/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Morfogênese , Neoplasias/metabolismo , Pele/crescimento & desenvolvimento , Pele/patologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proliferação de Células , Gefitinibe , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Quinazolinas/metabolismo , Pele/citologia , Pele/metabolismo
10.
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
11.
Blood ; 101(12): 4937-43, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12576323

RESUMO

The Janus kinase Jak1 has been implicated in tumor formation by the Abelson oncogene. In this study we show that loss of Jak1 does not affect in vitro transformation by v-abl as defined by the ability to induce cytokine-independent B-cell colony formation or establishment of B-cell lines. However, Jak1-deficient, v-abl-transformed cell lines were more tumorgenic than wild-type cells when transplanted subcutaneously into severe combined immunodeficient (SCID) mice or injected intravenously into nude mice. Jak1 deficiency was associated with a loss in the ability of interferon-gamma (IFN-gamma)to induce growth arrest and/or apoptosis of v-abl-transformed pre-B cells or tumor growth in SCID mice. Moreover, IFN-gamma mRNA could be detected in growing tumors, and tumor cells explanted from SCID mice had lost the ability to respond to IFN-gamma in 9 of 20 cases, whereas the response to interferon-alpha (IFN-alpha) remained intact. Importantly, a similar increase in tumorgenicity was observed when IFN-gamma-deficient cells were injected into SCID mice, identifying the tumor cell itself as the main source of IFN-gamma. These findings demonstrate that Jak1, rather than promoting tumorgenesis as previously proposed, is critical in mediating an intrinsic IFN-gamma-dependent tumor surveillance.


Assuntos
Vírus da Leucemia Murina de Abelson , Linfócitos B , Transformação Celular Neoplásica , Proteínas Tirosina Quinases/deficiência , Animais , Técnicas de Cultura , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Janus Quinase 1 , Fígado/embriologia , Camundongos , Camundongos Nus , Camundongos SCID , Transplante de Neoplasias , Hibridização de Ácido Nucleico , Proteínas Oncogênicas v-abl/genética , Proteínas Tirosina Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Biol Chem ; 277(36): 32490-7, 2002 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-12082090

RESUMO

The small G protein RAP1 and the kinase B-RAF have been proposed to link elevations of cAMP to activation of ERK/mitogen-activated protein (MAP) kinase. In order to delineate signaling pathways that link receptor-generated cAMP to the activation of MAP kinase, the human A(2A)-adenosine receptor, a prototypical G(s)-coupled receptor, was heterologously expressed in Chinese hamster ovary cells (referred as CHO-A(2A) cells). In CHO-A(2A) cells, the stimulation of the A(2A)-receptor resulted in an activation of RAP1 and formation of RAP1-B-RAF complexes. However, overexpression of a RAP1 GTPase-activating protein (RAP1GAP), which efficiently clamped cellular RAP1 in the inactive GDP-bound form, did not affect A(2A)-agonist-mediated MAP kinase stimulation. In contrast, the inhibitor of protein kinase A H89 efficiently suppressed A(2A)-agonist-mediated MAP kinase stimulation. Neither dynamin-dependent receptor internalization nor receptor-promoted shedding of matrix-bound growth factors accounted for A(2A)-receptor-dependent MAP kinase activation. PP1, an inhibitor of SRC family kinases, blunted both the A(2A)-receptor- and the forskolin-induced MAP kinase stimulation (IC(50) = 50 nm); this was also seen in PC12 cells, which express the A(2A)-receptor endogenously, and in NIH3T3 fibroblasts, in which cAMP causes MAP kinase stimulation. In the corresponding murine fibroblast cell line SYF, which lacks the ubiquitously expressed SRC family kinases SRC, YES, and FYN, forskolin barely stimulated MAP kinase; this reduction was reversed in cells in which c-SRC had been reintroduced. These findings show that activation of MAP kinase by cAMP requires a SRC family kinase that lies downstream of protein kinase A. A role for RAP1, as documented for the beta(2)-adrenergic receptor, is apparently contingent on receptor endocytosis.


Assuntos
AMP Cíclico/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Sulfonamidas , Proteínas rap1 de Ligação ao GTP/fisiologia , Quinases da Família src/fisiologia , Células 3T3 , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Animais , Células CHO , Linhagem Celular , Colforsina/farmacologia , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Dinaminas , Endocitose , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , GTP Fosfo-Hidrolases/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Vetores Genéticos , Glutationa Transferase/metabolismo , Humanos , Immunoblotting , Concentração Inibidora 50 , Isoproterenol/farmacologia , Isoquinolinas/farmacologia , Camundongos , Células PC12 , Fosforilação , Testes de Precipitina , Ligação Proteica , Ratos , Fatores de Tempo , Ativação Transcricional , Transfecção
13.
Mol Pharmacol ; 61(3): 628-36, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11854444

RESUMO

We have previously shown that alkyl-substituted amino acid derivatives directly activate G(i/o) proteins. N-Dodecyl-N(alpha),N(epsilon)-(bis-l-lysinyl)-l-lysine amide (FUB132) is a new representative of this class of compounds with increased efficacy. Here, we characterized the molecular mechanism of action of this class of compounds. FUB132 and its predecessor FUB86 were selective receptomimetics for G(i/o) because they stimulated the guanine nucleotide exchange reaction of purified G(i/o) as documented by an increased rate of GDP release, GTP gamma S binding, and GTP hydrolysis. In contrast to the receptomimetic peptide mastoparan, stimulation of G proteins by lipoamines required the presence of neither G beta gamma-dimers nor lipids. On the contrary, G beta gamma-dimers suppressed the stimulatory effect of FUB132. The stimulation of G(i/o) by lipoamines and by mastoparan was not additive. A peptide derived from the C terminus of G alpha(o3), but not a corresponding G alpha(q)-derived peptide, quenched the FUB132-induced activation of G alpha(o). In membranes prepared from human embryonic kidney 293 cells that stably expressed the G(i/o)-coupled human A(1)-adenosine receptor, lipoamines impeded high-affinity agonist binding. In contrast, antagonist binding was not affected. We conclude that alkyl-substituted amines target a site, most likely at the C terminus of G alpha(i/o)-subunits, that is also contacted by receptors. However, because G beta gamma-dimers blunt rather than enhance their efficacy, their mechanism of action differs fundamentally from that of a receptor. Thus, despite their receptomimetic effect in vitro, alkyl-substituted amines and related polyamines are poor direct G protein activators in vivo. In the presence of G beta gamma, they rather antagonize G protein-coupled receptor signaling.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Lisina/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Células HL-60 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Lisina/análogos & derivados , Peptídeos , Ensaio Radioligante , Transdução de Sinais/efeitos dos fármacos , Venenos de Vespas/farmacologia
14.
Naunyn Schmiedebergs Arch Pharmacol ; 365(1): 8-16, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11862328

RESUMO

The interface between receptors and G proteins can be considered as a drug target. Various classes of low molecular weight inhibitors have been identified that block the ability of receptors to interact with G proteins (e.g. peptides, suramin analogues and amphiphilic cations). Here we have tested if there are compounds that differentially affect the interaction of one receptor with two different (related) G protein alpha-subunits. Fusion proteins comprising the human A1-adenosine receptor and Galphai-1 (A1/Galphai-1) or Galphao (A1/Galphao) were expressed in HEK293 cells. Suramin analogues were screened for their ability to differentially affect high affinity binding of the agonist (-) N6-3-[125I](iodo-4-hydroxyphenylisopropyl) adenosine (IHPIA). One compound [NF326 = 8,8'-(carbonylbis-(imino-3,1-phenylenecarbonylimino))bis-(1-naphthol-3,6-disulfonic acid, disodium salt)] was identified that inhibited high affinity agonist binding to the fusion protein A1/Galphai-1 but modestly enhanced binding of IHPIA to A1/Galphao. This action was specific because NF326 did not affect antagonist binding to either fusion protein. In addition, it was unrelated to a difference in affinity of the receptor for the G protein fusion moiety because the stability of ternary complexes formed by IHPIA + A1/Galphai-1 and IHPIA + A1/Galphao) is comparable and because lowering the affinity of the receptor for the G protein (by introducing point mutations at cys351 of Galphai-1) enhanced the uncoupling effect of NF326. Finally, NF326 did not discriminate between a fusion protein comprising the alpha2A-adrenoceptor and Galphai-1 (alpha2A)/Galphai-1) or Galphao-1 (alpha2A)/Galphao-1); binding of the agonist [3H]UK14304 (bromoxidine) to both fusion proteins was inhibited over a comparable concentration range while binding of the antagonist [3H]yohimbine was unaffected. These observations are consistent with the interpretation that the contact sites that are formed between individual receptors and G proteins differ. These differences suffice to allow for selective disruption by G protein inhibitors of different classes. Using NF326 we show that the bulk of the A1-adenosine receptors in human cerebrocortical membranes interacts with Galphao rather than Galphai.


Assuntos
Encéfalo/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Antagonistas de Receptores Purinérgicos P1 , Receptores Purinérgicos P1/metabolismo , Suramina/análogos & derivados , Suramina/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Células COS , Relação Dose-Resposta a Droga , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP , Humanos , Suramina/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA