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












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38334646

RESUMEN

Protein aggregation is a predominant feature of many neurodegenerative diseases, including synucleinopathies, which are characterized by cellular inclusions containing α-Synuclein (αSyn) phosphorylated at serine 129 (pSer129). In the present study, we characterized the development of αSyn pre-formed fibril (PFF)-induced pSer129-αSyn pathology in F28tg mice overexpressing human wild-type αSyn, as well as in ex vivo organotypic cultures and in vitro primary cultures from the same mouse model. Concurrently, we collected cerebrospinal fluid (CSF) from mice and conditioned media from ex vivo and in vitro cultures and quantified the levels of neurofilament light chain (NFL), a biomarker of neurodegeneration. We found that the intra-striatal injection of PFFs induces the progressive spread of pSer129-αSyn pathology and microglial activation in vivo, as well as modest increases in NFL levels in the CSF. Similarly, PFF-induced αSyn pathology occurs progressively in ex vivo organotypic slice cultures and is accompanied by significant increases in NFL release into the media. Using in vitro primary hippocampal cultures, we further confirmed that pSer129-αSyn pathology and NFL release occur in a manner that correlates with the fibril dose and the level of the αSyn protein. Overall, we demonstrate that αSyn pathology is associated with NFL release across preclinical models of seeded αSyn aggregation and that the pharmacological inhibition of αSyn aggregation in vitro also significantly reduces NFL release.


Asunto(s)
Enfermedades Neurodegenerativas , Sinucleinopatías , Animales , Humanos , Ratones , alfa-Sinucleína/metabolismo , Filamentos Intermedios/metabolismo , Enfermedades Neurodegenerativas/patología , Agregado de Proteínas/fisiología
2.
Sci Signal ; 16(779): eabl4283, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-37014928

RESUMEN

The chemotactic G protein-coupled receptor GPR183 and its most potent endogenous oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) are important for immune cell positioning in secondary lymphoid tissues. This receptor-ligand pair is associated with various diseases, in some cases contributing favorably and in other cases adversely, making GPR183 an attractive target for therapeutic intervention. We investigated the mechanisms underlying GPR183 internalization and the role of internalization in the main biological function of the receptor, chemotaxis. We found that the C terminus of the receptor was important for ligand-induced internalization but less so for constitutive (ligand-independent) internalization. ß-arrestin potentiated ligand-induced internalization but was not required for ligand-induced or constitutive internalization. Caveolin and dynamin were the main mediators of both constitutive and ligand-induced receptor internalization in a mechanism independent of G protein activation. Clathrin-mediated endocytosis also contributed to constitutive GPR183 internalization in a ß-arrestin-independent manner, suggesting the existence of different pools of surface-localized GPR183. Chemotaxis mediated by GPR183 depended on receptor desensitization by ß-arrestins but could be uncoupled from internalization, highlighting an important biological role for the recruitment of ß-arrestin to GPR183. The role of distinct pathways in internalization and chemotaxis may aid in the development of GPR183-targeting drugs for specific disease contexts.


Asunto(s)
Arrestina , Arrestinas , Arrestina/metabolismo , Arrestinas/genética , Arrestinas/metabolismo , Ligandos , beta-Arrestinas/metabolismo , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , Endocitosis
3.
Commun Biol ; 4(1): 569, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980979

RESUMEN

Following the FDA-approval of the hematopoietic stem cell (HSC) mobilizer plerixafor, orally available and potent CXCR4 antagonists were pursued. One such proposition was AMD11070, which was orally active and had superior antagonism in vitro; however, it did not appear as effective for HSC mobilization in vivo. Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating ß-arrestin recruitment while fully antagonizing G protein. Consequently, while AMD11070 prevents the constitutive receptor internalization, plerixafor allows it and thereby decreases receptor expression. These findings are confirmed by the successful transfer of both ligands' binding sites and action to the related CXCR3 receptor. In vivo, plerixafor exhibits superior HSC mobilization associated with a dramatic reversal of the CXCL12 gradient across the bone marrow endothelium, which is not seen for AMD11070. We propose that the biased action of plerixafor is central for its superior therapeutic effect in HSC mobilization.


Asunto(s)
Bencilaminas/farmacología , Ciclamas/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Receptores CXCR4/metabolismo , Aminoquinolinas/metabolismo , Aminoquinolinas/farmacología , Animales , Bencimidazoles/metabolismo , Bencimidazoles/farmacología , Bencilaminas/metabolismo , Butilaminas/metabolismo , Butilaminas/farmacología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Ciclamas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Femenino , Factor Estimulante de Colonias de Granulocitos , Células HEK293 , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Preparaciones Farmacéuticas/metabolismo , Receptores CXCR3/efectos de los fármacos , Receptores CXCR3/metabolismo , Receptores CXCR4/efectos de los fármacos , beta-Arrestinas/efectos de los fármacos , beta-Arrestinas/metabolismo
4.
Eur J Pharmacol ; 887: 173440, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32745603

RESUMEN

Activation of the voltage-gated Kv7 channels holds therapeutic promise in several neurological and psychiatric disorders, including epilepsy, schizophrenia, and depression. Here, we present a pharmacological characterization of Lu AA41178, a novel, pan-selective Kv7.2-7.5 opener, using both in vitro assays and a broad range of in vivo assays with relevance to epilepsy, schizophrenia, and depression. Electrophysiological characterization in Xenopus oocytes expressing human Kv7.2-Kv7.5 confirmed Lu AA41178 as a pan-selective opener of Kv7 channels by significantly left-shifting the activation threshold. Additionally, Lu AA41178 was tested in vitro for off-target effects, demonstrating a clean Kv7-selective profile, with no impact on common cardiac ion channels, and no potentiating activity on GABAA channels. Lu AA41178 was evaluated across preclinical in vivo assays with relevance to neurological and psychiatric disorders. In the maximum electroshock seizure threshold test and PTZ seizure threshold test, Lu AA41178 significantly increased the seizure thresholds in mice, demonstrating anticonvulsant efficacy. Lu AA41178 demonstrated antipsychotic-like activity by reducing amphetamine-induced hyperlocomotion in mice as well as lowering conditioned avoidance responses in rats. In the mouse forced swim test, a model with antidepressant predictivity, Lu AA41178 significantly reduced immobility. Additionally, behavioral effects typically observed with Kv7 openers was also characterized. In vivo assays were accompanied by plasma and brain exposures, revealing minimum effective plasma levels <1000 ng/ml. Lu AA41178, a potent opener of neuronal Kv7 channels demonstrate efficacy in assays of epilepsy, schizophrenia and depression and might serve as a valuable tool for exploring the role of Kv7 channels in both neurological and psychiatric disorders.


Asunto(s)
Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Canal de Potasio KCNQ2/agonistas , Trastornos Mentales/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Canal de Potasio KCNQ2/metabolismo , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/psicología , Ratones , Ratones Endogámicos C57BL , Psicotrópicos/farmacología , Psicotrópicos/uso terapéutico , Ratas , Ratas Wistar , Convulsiones/metabolismo , Convulsiones/psicología , Resultado del Tratamiento , Xenopus laevis
5.
Front Cell Neurosci ; 14: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32116557

RESUMEN

The voltage-gated Kv7.2/Kv7.3 potassium channel is a critical regulator of neuronal excitability. It is strategically positioned at the axon initial segment (AIS) of neurons, where it effectively inhibits repetitive action potential firing. While the selective accumulation of Kv7.2/Kv7.3 channels at the AIS requires binding to the adaptor protein ankyrin G, it is currently unknown if additional molecular mechanisms contribute to the localization and fine-tuning of channel numbers at the AIS. Here, we utilized a chimeric approach to pinpoint regions within the Kv7.3 C-terminal tail with an impact upon AIS localization. This strategy identified two domains with opposing effects upon the AIS localization of Kv7.3 chimeras expressed in cultured hippocampal neurons. While a membrane proximal domain reduced AIS localization of Kv7.3 chimeras, helix D increased and stabilized chimera AIS localization. None of the identified domains were required for AIS localization. However, the domains modulated the relative efficiency of the localization raising the possibility that the two domains contribute to the regulation of Kv7 channel numbers and nanoscale organization at the AIS.

6.
Br J Pharmacol ; 174(13): 2031-2042, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28369721

RESUMEN

BACKGROUND AND PURPOSE: The GPCR Epstein-Barr virus-induced gene 2 (EBI2, also known as GPR183) is activated by oxysterols and plays a pivotal role in the regulation of B cell migration during immune responses. While the molecular basis of agonist binding has been addressed in several studies, the concept of biased agonism of the EBI2 receptor has not been explored. EXPERIMENTAL APPROACH: We investigated the effects of the EBI2 endogenous agonist 7α,25-dihydroxycholesterol (7α,25-OHC) on G protein-dependent and -independent pathways as well as sodium ion allosterism using site-directed mutagenesis and functional studies. Moreover, we generated a homology model of the EBI2 receptor to investigate the structural basis of the allosteric modulation by sodium. KEY RESULTS: Residue N114, located in the middle of transmembrane-III at position III:11/3.35, was found to function as an efficacy switch. Thus, substituting N114 with an alanine (N114A) completely abolished heterotrimeric G protein subunit Gi α activation by 7α,25-OHC even though the specific binding of [3 H]-7α,25-OHC increased. In contrast, the N114A mutant was still able to recruit ß-arrestin and even had an enhanced potency (18.7-fold) compared with EBI2 wild type. Sodium had a negative allosteric effect on oxysterol binding that was mediated via N114, verifying the key role of N114. This was further supported by molecular modelling of the ion binding site based on a EBI2 receptor homology model. CONCLUSIONS AND IMPLICATIONS: Collectively, our data point to N114 as a key residue for EBI2 signalling controlling the balance between G protein-dependent and -independent pathways and facilitating sodium binding.


Asunto(s)
Hidroxicolesteroles/farmacología , Receptores Acoplados a Proteínas G/agonistas , Regulación Alostérica/efectos de los fármacos , Animales , Unión Competitiva/efectos de los fármacos , Células CHO , Células Cultivadas , Cricetulus , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Sodio/farmacología , Relación Estructura-Actividad
7.
Cell Signal ; 30: 9-18, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27865873

RESUMEN

G protein-coupled receptors (GPCRs) constitute a large protein family of seven transmembrane (7TM) spanning proteins that regulate multiple physiological functions. GPR87 is overexpressed in several cancers and plays a role in tumor cell survival. Here, the basal activity of GPR87 was investigated in transiently transfected HEK293 cells, revealing ligand-independent coupling to Gαi, Gαq and Gα12/13. Furthermore, GPR87 showed a ligand-independent G protein-dependent activation of the downstream transcription factors CREB, NFκB, NFAT and SRE. In tetracycline-induced Flp-In T-Rex-293 cells, GPR87 induced cell clustering presumably through Gα12/13 coupling. In a foci formation assay using retrovirally transduced NIH3T3 cells, GPR87 showed a strong in vitro transforming potential, which correlated to the in vivo tumor induction in nude mice. Importantly, we demonstrate that the transforming potential of GPR87 was correlated to the receptor signaling, as the signaling-impaired mutant R139A (Arg in the conserved "DRY"-motif at the bottom of transmembrane helix 3 of GPR87 substituted to Ala) showed a lower in vitro cell transformation potential. Furthermore, R139A lost the ability to induce cell clustering. In summary, we show that GPR87 is active through several signaling pathways and that the signaling activity is linked to the receptor-induced cell transformation and clustering. The robust surface expression of GPR87 and general high druggability of GPCRs make GPR87 an attractive future anticancer target for drugs that - through inhibition of the receptor signaling - will inhibit its transforming properties.


Asunto(s)
Carcinogénesis/metabolismo , Carcinogénesis/patología , Proteínas de Unión al GTP/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Animales , Células COS , Membrana Celular/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Chlorocebus aethiops , AMP Cíclico/metabolismo , Femenino , Células HEK293 , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Ligandos , Lisofosfolípidos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transfección , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo
8.
Blood ; 129(7): 866-878, 2017 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-28003273

RESUMEN

Human and mouse chronic lymphocytic leukemia (CLL) develops from CD5+ B cells that in mice and macaques are known to define the distinct B1a B-cell lineage. B1a cells are characterized by lack of germinal center (GC) development, and the B1a cell population is increased in mice with reduced GC formation. As a major mediator of follicular B-cell migration, the G protein-coupled receptor Epstein-Barr virus-induced gene 2 (EBI2 or GPR183) directs B-cell migration in the lymphoid follicles in response to its endogenous ligands, oxysterols. Thus, upregulation of EBI2 drives the B cells toward the extrafollicular area, whereas downregulation is essential for GC formation. We therefore speculated whether increased expression of EBI2 would lead to an expanded B1 cell subset and, ultimately, progression to CLL. Here, we demonstrate that B-cell-targeted expression of human EBI2 (hEBI2) in mice reduces GC-dependent immune responses, reduces total immunoglobulin M (IgM) and IgG levels, and leads to increased proliferation and upregulation of cellular oncogenes. Furthermore, hEBI2 overexpression leads to an abnormally expanded CD5+ B1a B-cell subset (present as early as 4 days after birth), late-onset lymphoid cancer development, and premature death. These findings are highly similar to those observed in CLL patients and identify EBI2 as a promoter of B-cell malignancies.


Asunto(s)
Linfocitos B/patología , Centro Germinal/patología , Leucemia Linfocítica Crónica de Células B/genética , Linfoma/genética , Receptores Acoplados a Proteínas G/genética , Regulación hacia Arriba , Animales , Linfocitos B/inmunología , Antígenos CD5/análisis , Antígenos CD5/inmunología , Regulación Neoplásica de la Expresión Génica , Centro Germinal/citología , Centro Germinal/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Linfoma/inmunología , Linfoma/patología , Ratones , Receptores Acoplados a Proteínas G/inmunología
9.
J Neurosci ; 36(7): 2261-6, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26888935

RESUMEN

The voltage-gated K(+) channels Kv7.2 and Kv7.3 are located at the axon initial segment (AIS) and exert strong control over action potential generation. Therefore, changes in their localization or cell surface numbers are likely to influence neuronal signaling. However, nothing is known about the cell surface dynamics of Kv7.2/7.3 at steady state or during short-term neuronal stimulation. This is primarily attributable to their membrane topology, which hampers extracellular epitope tagging. Here we circumvent this limitation by fusing an extra phluorin-tagged helix to the N terminus of human Kv7.3. This seven transmembrane chimera, named super ecliptic phluorin (SEP)-TAC-7.3, functions and traffics as a wild-type (WT) channel. We expressed SEP-TAC-7.3 in dissociated rat hippocampal neurons to examine the lateral mobility, surface numbers, and localization of AIS Kv7.2/7.3 heteromers using live imaging. We discovered that they are extraordinarily stable and exhibit a very low surface mobility both during steady state and neuronal stimulation. In the latter case, we also found that neither localization nor cell surface numbers were changed. However, at high glutamate loads, we observed a rapid irreversible endocytosis of Kv7.2/7.3, which required the activation of NR2B-containing NMDA receptors, Ca(2+) influx, and calpain activation. This excitotoxic mechanism may be specific to ankyrin G-bound AIS proteins because Nav1.2 channels, but not AIS GABAA receptors, were also endocytosed. In conclusion, we have, for the first time, characterized the cell surface dynamics of a full-length Kv7 channel using a novel chimeric strategy. This approach is likely also applicable to other Kv channels and thus of value for the additional characterization of this ion channel subfamily. SIGNIFICANCE STATEMENT: The voltage-gated K(+) channels Kv7.2 and Kv7.3 exert strong control over action potential generation, but little is known about their cell surface dynamics. Using a novel phluorin-based approach, we here show that these channels are highly stable at steady state and different types of neuronal stimulation. However, at high glutamate loads, they undergo a rapid calpain-dependent endocytosis that likely represents an early response during excitotoxic states.


Asunto(s)
Axones/metabolismo , Calpaína/metabolismo , Regulación hacia Abajo/genética , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Ancirinas/genética , Axones/ultraestructura , Señalización del Calcio/genética , Quimera/genética , Femenino , Humanos , Canal de Potasio KCNQ2/ultraestructura , Canal de Potasio KCNQ3/ultraestructura , Masculino , Ratones , Proteínas del Tejido Nervioso/ultraestructura , Técnicas de Placa-Clamp , Embarazo , Ratas , Receptores de Superficie Celular/metabolismo , Receptores de GABA-A/genética , Receptores de N-Metil-D-Aspartato/genética
10.
Eur J Immunol ; 44(7): 1904-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24810762

RESUMEN

The seven transmembrane G protein-coupled receptor Epstein-Barr virus (EBV) induced gene 2 (EBI2; also known as GPR183) was identified in 1993 on the basis of its substantial upregulation in EBV-infected cells. It is primarily expressed in lymphoid cells; most abundantly in B cells. EBI2 is central for the positioning of B cells within the lymphoid organs, a process that is regulated in part by a chemotactic gradient formed by the endogenous lipid agonists, and in part by a fine-tuned regulation of EBI2 cell surface expression. The most potent endogenous EBI2 agonist is 7α, 25-dihydroxyxcholesterol (7α,25-OHC), yet many structurally related oxysterols can bind to an EBI2 pocket that is defined by the upper parts of the transmembrane helices and extracellular receptor regions. EBI2 signals via Gαi, as well as via G protein-independent pathways like ß-arrestin recruitment. The concerted action of these pathways leads to cell migration. By genetically interfering with its up- and downregulation, EBI2 was also recently shown to induce cell proliferation, an action that could be inhibited by small molecule antagonists. Here, we focus on the oxysterol-EBI2 axis in immune control, including its role in the EBV life cycle. We also summarize the structural and functional properties of EBI2 interaction with oxysterol agonists and small molecule antagonists and discuss EBI2 as therapeutic target for diseases of the immune system.


Asunto(s)
Colesterol/metabolismo , Infecciones por Virus de Epstein-Barr/inmunología , Hidroxicolesteroles/metabolismo , Sistema Inmunológico/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/fisiología , Animales , Humanos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química
11.
FEBS Open Bio ; 3: 156-60, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23772388

RESUMEN

The Epstein-Barr virus induced gene 2 (EBI2) was recently identified as the first oxysterol-activated 7TM receptor. EBI2 is essential for B cell trafficking within lymphoid tissues and thus the humoral immune response in general. Here we characterize the antagonism of the non-peptide molecule GSK682753A, which blocks oxysterol-induced G-protein activation, ß-arrestin recruitment and B-cell chemotaxis. We furthermore demonstrate that activation triggers pertussis toxin-sensitive MAP kinase phosphorylation, which is also inhibited by GSK682753A. Thus, EBI2 signalling in B cells mediates key phenotypic functions via signalling pathways amenable to manipulation providing additional therapeutic options for inhibiting EBI2 activity.

12.
J Biol Chem ; 287(42): 35470-35483, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22875855

RESUMEN

Oxysterols are oxygenated cholesterol derivates that are emerging as a physiologically important group of molecules. Although they regulate a range of cellular processes, only few oxysterol-binding effector proteins have been identified, and the knowledge of their binding mode is limited. Recently, the family of G protein-coupled seven transmembrane-spanning receptors (7TM receptors) was added to this group. Specifically, the Epstein-Barr virus-induced gene 2 (EBI2 or GPR183) was shown to be activated by several oxysterols, most potently by 7α,25-dihydroxycholesterol (7α,25-OHC). Nothing is known about the binding mode, however. Using mutational analysis, we identify here four key residues for 7α,25-OHC binding: Arg-87 in TM-II (position II:20/2.60), Tyr-112 and Tyr-116 (positions III:09/3.33 and III:13/3.37) in TM-III, and Tyr-260 in TM-VI (position VI:16/6.51). Substituting these residues with Ala and/or Phe results in a severe decrease in agonist binding and receptor activation. Docking simulations suggest that Tyr-116 interacts with the 3ß-OH group in the agonist, Tyr-260 with the 7α-OH group, and Arg-87, either directly or indirectly, with the 25-OH group, although nearby residues likely also contribute. In addition, Tyr-112 is involved in 7α,25-OHC binding but via hydrophobic interactions. Finally, we show that II:20/2.60 constitutes an important residue for ligand binding in receptors carrying a positively charged residue at this position. This group is dominated by lipid- and nucleotide-activated receptors, here exemplified by the CysLTs, P2Y12, and P2Y14. In conclusion, we present the first molecular characterization of oxysterol binding to a 7TM receptor and identify position II:20/2.60 as a generally important residue for ligand binding in certain 7TM receptors.


Asunto(s)
Dominio Catalítico , Hidroxicolesteroles/química , Simulación del Acoplamiento Molecular , Receptores Acoplados a Proteínas G/química , Sustitución de Aminoácidos , Células HEK293 , Humanos , Hidroxicolesteroles/metabolismo , Mutación Missense , Unión Proteica , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
13.
PLoS One ; 6(9): e24644, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931793

RESUMEN

BACKGROUND: The melanocortin 1 receptor (MC1R) constitutes a key regulator of melanism. Consequently, many naturally-occurring MC1R mutations are associated with a change in color. An example is the Glu-to-Lys substitution found at position II:20/2.60 in the top of transmembrane helix II which has been identified in melanic mice and several other species. This mutation induces a pronounced increase in MC1R constitutive activity suggesting a link between constitutive activity and melanism which is corroborated by the attenuation of α-melanocyte stimulating hormone (αMSH) induced activation. However, the mechanism by which the mutation induces constitutive activity is currently not known. METHODOLOGY/PRINCIPAL FINDINGS: Here we characterize the constitutive activity, cell surface expression and internalization of the mouse mutant, Mc1r E92K. As previously reported, only positively charged residues at position II:20/2.60 induced an increase in constitutive activity as measured by cAMP accumulation and CREB activation. Furthermore, the mutation induced a constitutive recruitment of ß-arrestin. This phenomenon is only observed in MC1R, however, as the equivalent mutations in MC2-5R had no effect on receptor signaling. Interestingly, the mutation did not induce constitutive ERK1/2 phosphorylation or increase the internalization rate indicating the constitutive activity to be biased. Finally, to identify regions of importance for the increased constitutive activity of Mc1r E92K, we employed a chimeric approach and identified G102 and L110 in the extracellular loop 1 to be selectively important for the constitutive activity as this, but not αMSH-mediated activation, was abolished upon Ala substitution. CONCLUSIONS/SIGNIFICANCE: It is concluded that the E92K mutation induces an active conformation distinct from that induced by αMSH and that the extracellular loop 1 is involved in maintaining this conformational state. In turn, the results suggest that in MC1R, which lacks an extracellular loop 2, the first extracellular loop may play a more prominent role during receptor activation than in general.


Asunto(s)
Arrestina/metabolismo , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Animales , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Receptor de Melanocortina Tipo 1/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , alfa-MSH/genética , alfa-MSH/metabolismo
14.
J Biol Chem ; 286(33): 29292-29302, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21673108

RESUMEN

The Epstein-Barr virus-induced receptor 2 (EBI2) is a constitutively active seven-transmembrane receptor, which was recently shown to orchestrate the positioning of B cells in the follicle. To date, no ligands, endogenously or synthetic, have been identified that modulate EBI2 activity. Here we describe an inverse agonist, GSK682753A, which selectively inhibited the constitutive activity of EBI2 with high potency and efficacy. In cAMP-response element-binding protein-based reporter and guanosine 5'-3-O-(thio)triphosphate (GTPγS) binding assays, the potency of this compound was 2.6-53.6 nm, and its inhibitory efficacy was 75%. In addition, we show that EBI2 constitutively activated extracellular signal-regulated kinase (ERK) in a pertussis toxin-insensitive manner. Intriguingly, GSK682753A inhibited ERK phosphorylation, GTPγS binding, and cAMP-response element-binding protein activation with similar potency. Overexpression of EBI2 profoundly potentiated antibody-stimulated ex vivo proliferation of murine B cells compared with WT cells, whereas this was equivalently reduced for EBI2-deficient B cells. Inhibition of EBI2 constitutive activity suppressed the proliferation in all cases. Importantly, the suppression was of much higher potency (32-fold) in WT or EBI2-overexpressing B cells compared with EBI2-deficient counterparts. Finally, we screened GSK682753A against an EBI2 mutant library to determine putative molecular binding determinants in EBI2. We identified Phe(111) at position III:08/3.32 as being crucial for GSK682753A inverse agonism because Ala substitution resulted in a >500-fold decrease in IC(50). In conclusion, we present the first ligand targeting EBI2. In turn, this molecule provides a useful tool for further characterization of EBI2 as well as serving as a potent lead compound.


Asunto(s)
Linfocitos B/metabolismo , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Oxazoles/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Compuestos de Espiro/farmacología , Animales , Linfocitos B/citología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Compuestos Heterocíclicos de 4 o más Anillos/química , Humanos , Ratones , Ratones Mutantes , Oxazoles/química , Fosforilación/efectos de los fármacos , Fosforilación/genética , Receptores Acoplados a Proteínas G/genética , Elementos de Respuesta/fisiología , Compuestos de Espiro/química
15.
Curr Top Med Chem ; 11(6): 618-28, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21261596

RESUMEN

7TM receptors constitute one of the largest superfamilies of proteins in the human genome. They are involved in a large number of physiological and pathological processes in the human body and thus represent major and important drug targets for the pharmaceutical industry. Although the majority have been deorphanized, many remain orphan, and these orphan receptors constitute a large pool of potential drug targets. This review focuses on one of these orphan targets, the Epstein-Barr Virus-induced receptor 2, EBI2 (or GPR183), together with two structurally related receptors, GPR17 and GPR18. The pharmacology and "druggability" of these three receptors is reviewed through a thorough description of their structural and functional properties and in vivo biology together with a status of currently available ligands for these receptors.


Asunto(s)
Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Conformación Proteica , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/genética
16.
Trends Pharmacol Sci ; 31(12): 567-74, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20870300

RESUMEN

From the deep part of the main ligand-binding crevice, a minor, often shallower pocket extends between the extracellular ends of transmembrane domains (TM)-I, II, III and VII of 7TM receptors. This minor binding pocket is defined by a highly conserved kink in TM-II that is induced by a proline residue located in one of two adjacent positions. Here we argue that this minor binding pocket is important for receptor activation. Functional coupling of the receptors seems to be mediated through the hydrogen bond network located between the intracellular segments of these TMs, with the allosteric interface between TM-II and TM-VII being of particular significance. Importantly, the minor binding pocket, especially the proline-kink in TM-II, is involved in G protein versus arrestin pathway-biased signaling, for example in the angiotensin AT1 system. Consequently, this pocket could be specifically targeted in the development of functionally biased drugs.


Asunto(s)
Sitios de Unión , Ligandos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos , Modelos Moleculares , Estructura Cuaternaria de Proteína , Transducción de Señal
17.
Mol Pharmacol ; 74(4): 1008-21, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18628402

RESUMEN

The Epstein-Barr induced receptor 2 (EBI2) is a lymphocyte-expressed orphan seven transmembrane-spanning (7TM) receptor that signals constitutively through Galphai, as shown, for instance by guanosine 5'-O-(3-thio)triphosphate incorporation. Two regions of importance for the constitutive activity were identified by a systematic mutational analysis of 29 residues in EBI2. The cAMP response element-binding protein transcription factor was used as a measure of receptor activity and was correlated to the receptor surface expression. PheVI:13 (Phe257), and the neighboring CysVI:12 (Cys256), in the conserved CW/FxP motif in TM 6, acted as negative regulators as Ala substitutions at these positions increased the constitutive activity 5.7- and 2.3-fold, respectively, compared with EBI2 wild type (wt). In contrast, ArgII:20 (Arg87) in TM-2 acted as a positive regulator, as substitution to Ala, but not to Lys, decreased the constitutive activity more than 7-fold compared with wt EBI2. IleIII:03 (Ile106) is located only 4 A from ArgII:20, and a favorable electrostatic interaction with ArgII:20 was created by introduction of Glu in III:03, given that the activity increased to 4.4-fold of that wt EBI2. It is noteworthy that swapping these charges by introduction of Glu in II:20 and Arg in III:03 resulted in a 2.7-fold increase compared with wt EBI2, thereby rescuing the two signaling-deficient single mutations, which exhibited a 3.8- to 4.5-fold decrease in constitutive activity. The uncovering of these molecular mechanisms for EBI2 activation is important from a drug development point of view, in that it may facilitate the rational design and development of small-molecule inverse agonists against EBI2 of putative importance as antiviral- or immune modulatory therapy.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Alanina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Aminoácidos Aromáticos/metabolismo , Línea Celular , Membrana Celular/metabolismo , Colforsina/farmacología , Secuencia Conservada , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/análisis , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Herpesvirus Humano 4/metabolismo , Humanos , Enlace de Hidrógeno , Riñón/citología , Luciferasas/metabolismo , Linfocitos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Electricidad Estática , Transfección
18.
J Virol ; 82(4): 1884-98, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18057236

RESUMEN

The murine cytomegalovirus (MCMV) M33 gene is conserved among all betaherpesviruses and encodes a homologue of seven-transmembrane receptors (7TMR) with the capacity for constitutive signaling. Previous studies have demonstrated that M33 is important for MCMV dissemination to or replication within the salivary glands. In this study, we probed N- and C-terminal regions of M33 as well as known 7TMR signature motifs in transmembrane (TM) II and TM III to determine the impact on cell surface expression, constitutive signaling, and in vivo phenotype. The region between amino acids R(340) and A(353) of the C terminus was found to be important for CREB- and NFAT-mediated signaling, although not essential for phosphatidylinositol turnover. Tagging or truncation of the N terminus of M33 resulted in loss of cell surface expression. Within TM II, an F79D mutation abolished constitutive signaling, demonstrating a role, as in other cellular and viral 7TMR, of TM II in receptor activation. In TM III, the arginine (but not the asparagine) residue of the NRY motif (the counterpart of the common DRY motif in cellular 7TMR) was found to be essential for constitutive signaling. Selected mutations incorporated into recombinant MCMV showed that disruption of constitutive signaling for a viral 7TMR homologue resulted in a reduced capacity to disseminate to or replicate in the salivary glands. In addition, HCMV UL33 was found to partially compensate for the lack of M33 in vivo, suggesting conserved biological roles of the UL33 gene family.


Asunto(s)
Muromegalovirus/fisiología , Receptores de Quimiocina/química , Receptores de Quimiocina/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Secuencia Conservada , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ratones , Muromegalovirus/genética , Factores de Transcripción NFATC/metabolismo , Fosfatidilinositoles/metabolismo , Mutación Puntual , Estructura Terciaria de Proteína , Receptores de Quimiocina/genética , Glándulas Salivales/virología , Transducción de Señal , Proteínas Virales/genética , Replicación Viral
19.
Mol Pharmacol ; 70(6): 1892-901, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16971553

RESUMEN

The extracellular part of transmembrane segment V (TM-V) is expected to be involved in the activation process of 7TM receptors, but its role is far from clear. Here, we study the highly constitutively active CXC-chemokine receptor encoded by human herpesvirus 8 (ORF74-HHV8), in which a metal ion site was introduced at the extracellular end of TM-V by substitution of two arginines at positions V:01 and V:05 with histidines [R208H; R212H]. The metal ion site conferred high-potency inverse agonist properties (EC(50), 1.7 microM) to Zn(II) in addition to agonist and allosteric enhancing properties at concentrations >10 microM. The chemokine interaction with [R208H;R212H]-ORF74 was altered compared with wild-type ORF74-HHV8 with decreased agonist (CXCL1/GROalpha) potency (84-fold), affinity (5.8- and 136-fold in competition against agonist and inverse agonist, respectively), and binding capacity (B(max); 25-fold). Zn(II) in activating concentrations (100 microM) acted as an allosteric enhancer as it increased the B(max) (7.1-fold), the potency (9.9-fold), the affinity (1.7- and 6.1-fold in competition against agonist and inverse agonist, respectively), and the efficacy (2.5-fold) of CXCL1/GROalpha. The activating properties of Zn(II) were not due to a metal ion site between the ligand and the receptor because CXCL1/GROalpha analogs in which the putative metal-ion binding residues had been substituted-[H19A] and [H34A]-acted like wild-type CXCL1/GROalpha. Based on the complex action of Zn(II) and on the chemokine interaction for [R208H;R212H]-ORF74, we conclude that the extracellular end of TM-V is important for the activation of this CXC-chemokine receptor.


Asunto(s)
Ingeniería de Proteínas , Receptores de Superficie Celular/química , Secuencia de Aminoácidos , Secuencia de Bases , Unión Competitiva , Dicroismo Circular , Clonación Molecular , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/genética , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Superficie Celular/genética
20.
J Biol Chem ; 281(19): 13199-13208, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16540462

RESUMEN

Epstein-Barr virus (EBV)-induced receptor 2 (EBI2) is an orphan seven-transmembrane (7TM) receptor originally identified as the most up-regulated gene (>200-fold) in EBV-infected cells. Here we show that EBI2 signals with constitutive activity through Galpha(i) as determined by a receptor-mediated inhibition of forskolin-induced cAMP production and an induction of the serum response element-driven transcriptional activity in a pertussis toxin-sensitive manner. Galpha(s) and Galpha(q) were not activated constitutively as determined by the lack of cAMP production, the lack of inositol phosphate turnover, and the lack of activities of the transcription factors: cAMP response element-binding protein and nuclear factor-kappaB. Immunohistochemistry and confocal microscopy of FLAG- and green fluorescent protein-tagged EBI2 revealed cell-surface expression. A putative N-terminal truncated version of EBI2, delta4-EBI2, showed similar expression and signaling through Galpha(i) as full-length EBI2. By using a 32P-labeled EBI2 probe we found a very high expression in lymphoid tissue (spleen and lymph node) and peripheral blood mononuclear cells and a high expression in lung tissue. Real-time PCR of EBV-infected cells showed high expression of EBI2 during latent and lytic infection, in contrast to the EBV-encoded 7TM receptor BILF1, which was induced during lytic infection. EBI2 clustered with the orphan GPR18 by alignment analysis as well as by close proximity in the chromosomal region 13q32.3. Based on the constitutive signaling and cellular expression pattern of EBI2, it is suggested that it may function in conjunction with BILF1 in the reprogramming of the cell during EBV infection.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Humanos , Células Asesinas Naturales/metabolismo , Linfocitos/metabolismo , Monocitos/metabolismo , Isoformas de Proteínas , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...