Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(11): e2308570121, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38442170

RESUMO

Cytokinesis is the last step of cell division and is regulated by the small GTPase RhoA. RhoA activity is required for all steps of cytokinesis, including prior to abscission when daughter cells are ultimately physically separated. Like germ cells in all animals, the Caenorhabditis elegans embryonic germline founder cell initiates cytokinesis but does not complete abscission, leaving a stable intercellular bridge between the two daughter cells. Here, we identify and characterize C. elegans OSGN-1 as a cytokinetic regulator that promotes RhoA activity during late cytokinesis. Sequence analyses and biochemical reconstitutions reveal that OSGN-1 is a flavin-containing monooxygenase (MO). Genetic analyses indicate that the MO activity of OSGN-1 is required to maintain active RhoA at the end of cytokinesis in the germline founder cell and to stabilize the intercellular bridge. Deletion of OSGIN1 in human cells results in an increase in binucleation as a result of cytokinetic furrow regression, and this phenotype can be rescued by expressing a catalytically active form of C. elegans OSGN-1, indicating that OSGN-1 and OSGIN1 are functional orthologs. We propose that OSGN-1 and OSGIN1 are conserved MO enzymes required to maintain RhoA activity at the intercellular bridge during late cytokinesis and thus favor its stability, enabling proper abscission in human cells and bridge stabilization in C. elegans germ cells.


Assuntos
Citocinese , Dermatite , Oxigenases , Animais , Humanos , Citocinese/genética , Caenorhabditis elegans/genética , Divisão Celular
2.
Development ; 148(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34195824

RESUMO

The C. elegans germline is organized as a syncytium in which each germ cell possesses an intercellular bridge that is maintained by a stable actomyosin ring and connected to a common pool of cytoplasm, termed the rachis. How germ cells undergo cytokinesis while maintaining this syncytial architecture is not completely understood. Here, we use live imaging to characterize primordial germ cell (PGC) division in C. elegans first-stage larvae. We show that each PGC possesses a stable intercellular bridge that connects it to a common pool of cytoplasm, which we term the proto-rachis. We further show that the first PGC cytokinesis is incomplete and that the stabilized cytokinetic ring progressively moves towards the proto-rachis and eventually integrates with it. Our results support a model in which the initial expansion of the C. elegans syncytial germline occurs by incomplete cytokinesis, where one daughter germ cell inherits the actomyosin ring that was newly formed by stabilization of the cytokinetic ring, while the other inherits the pre-existing stable actomyosin ring. We propose that such a mechanism of iterative cytokinesis incompletion underpins C. elegans germline expansion and maintenance.


Assuntos
Caenorhabditis elegans/citologia , Citocinese/fisiologia , Células Germinativas/citologia , Citoesqueleto de Actina/fisiologia , Actomiosina/fisiologia , Animais , Citoplasma/fisiologia , Células Gigantes/fisiologia
3.
J Cell Physiol ; 2022 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-36576983

RESUMO

Extracellular signal-regulated kinase 3 (ERK3) is a poorly characterized member of the mitogen-activated protein (MAP) kinase family. Functional analysis of the ERK3 signaling pathway has been hampered by a lack of knowledge about the substrates and downstream effectors of the kinase. Here, we used large-scale quantitative phosphoproteomics and targeted gene silencing to identify direct ERK3 substrates and gain insight into its cellular functions. Detailed validation of one candidate substrate identified the gelsolin/villin family member supervillin (SVIL) as a bona fide ERK3 substrate. We show that ERK3 phosphorylates SVIL on Ser245 to regulate myosin II activation and cytokinesis completion in dividing cells. Depletion of SVIL or ERK3 leads to increased cytokinesis failure and multinucleation, a phenotype rescued by wild type SVIL but not by the non-phosphorylatable S245A mutant. Our results unveil a new function of the atypical MAP kinase ERK3 in cell division and the regulation of cell ploidy.

4.
Proc Natl Acad Sci U S A ; 113(16): 4524-9, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27071102

RESUMO

G protein-coupled receptors (GPCRs) are known to initiate a plethora of signaling pathways in vitro. However, it is unclear which of these pathways are engaged to mediate physiological responses. Here, we examine the distinct roles of Gq/11-dependent signaling and receptor phosphorylation-dependent signaling in bronchial airway contraction and lung function regulated through the M3-muscarinic acetylcholine receptor (M3-mAChR). By using a genetically engineered mouse expressing a G protein-biased M3-mAChR mutant, we reveal the first evidence, to our knowledge, of a role for M3-mAChR phosphorylation in bronchial smooth muscle contraction in health and in a disease state with relevance to human asthma. Furthermore, this mouse model can be used to distinguish the physiological responses that are regulated by M3-mAChR phosphorylation (which include control of lung function) from those responses that are downstream of G protein signaling. In this way, we present an approach by which to predict the physiological/therapeutic outcome of M3-mAChR-biased ligands with important implications for drug discovery.


Assuntos
Brônquios/metabolismo , Músculo Liso/metabolismo , Receptor Muscarínico M3/metabolismo , Transdução de Sinais/fisiologia , Animais , Brônquios/citologia , Humanos , Camundongos , Camundongos Knockout , Músculo Liso/citologia , Fosforilação/fisiologia , Receptor Muscarínico M3/genética
5.
Methods ; 92: 11-8, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25962643

RESUMO

Ligand-biased signaling is starting to have significant impact on drug discovery programs in the pharmaceutical industry and has reinvigorated our understanding of pharmacological efficacy. As such, many investigators and screening campaigns are now being directed at a larger section of the signaling responses downstream of an individual G protein-coupled receptor. Many biosensor-based platforms have been developed to capture signaling signatures. Despite our growing ability to use such signaling signatures, we remain hampered by the fact that signaling signatures may be particular to an individual cell type and thus our platforms may not be portable from cell to cell, necessitating further cell-specific biosensor development. Here, we provide a complementary strategy based on capturing receptor-proximal conformational profiles using intra-molecular BRET-based sensors composed of a Renilla luciferase donor engineered into the carboxy-terminus and CCPGCC motifs which bind fluorescent hairpin arsenical dyes engineered into different positions in intracellular loop 3 of FP, the receptor for PGF2α. We discuss the design and optimization of such sensors for orthosteric and allosteric ligands.


Assuntos
Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Técnicas Biossensoriais/métodos , Desenho de Fármacos , Corantes Fluorescentes/síntese química , Receptores Acoplados a Proteínas G/química , Antagonistas do Receptor A2 de Adenosina/síntese química , Antagonistas do Receptor A2 de Adenosina/metabolismo , Sequência de Aminoácidos , Corantes Fluorescentes/metabolismo , Células HEK293 , Humanos , Luciferases de Renilla/síntese química , Luciferases de Renilla/metabolismo , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Receptor A2A de Adenosina/análise , Receptor A2A de Adenosina/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
6.
J Biol Chem ; 290(5): 3137-48, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25512374

RESUMO

The angiotensin II type I (AT1R) and the prostaglandin F2α (PGF2α) F prostanoid (FP) receptors are both potent regulators of blood pressure. Physiological interplay between AT1R and FP has been described. Abdominal aortic ring contraction experiments revealed that PGF2α-dependent activation of FP potentiated angiotensin II-induced contraction, whereas FP antagonists had the opposite effect. Similarly, PGF2α-mediated vasoconstriction was symmetrically regulated by co-treatment with AT1R agonist and antagonist. The underlying canonical Gαq signaling via production of inositol phosphates mediated by each receptor was also regulated by antagonists for the other receptor. However, binding to their respective agonists, regulation of receptor-mediated MAPK activation and vascular smooth muscle cell growth were differentially or asymmetrically regulated depending on how each of the two receptors were occupied by either agonist or antagonist. Physical interactions between these receptors have never been reported, and here we show that AT1R and FP form heterodimeric complexes in both HEK 293 and vascular smooth muscle cells. These findings imply that formation of the AT1R/FP dimer creates a novel allosteric signaling unit that shows symmetrical and asymmetrical signaling behavior, depending on the outcome measured. AT1R/FP dimers may thus be important in the regulation of blood pressure.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Prostaglandina/metabolismo , Regulação Alostérica/fisiologia , Animais , Células Cultivadas , Células HEK293 , Humanos , Ratos , Transdução de Sinais/fisiologia
7.
J Biol Chem ; 285(33): 25624-36, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20551320

RESUMO

The prostaglandin F2alpha (PGF2alpha) receptor (FP) is a key regulator of parturition and a target for pharmacological management of preterm labor. However, an incomplete understanding of signaling pathways regulating myometrial contraction hinders the development of improved therapeutics. Here we used a peptidomimetic inhibitor of parturition in mice, PDC113.824, whose structure was based on the NH(2)-terminal region of the second extracellular loop of FP receptor, to gain mechanistic insight underlying FP receptor-mediated cell responses in the context of parturition. We show that PDC113.824 not only delayed normal parturition in mice but also that it inhibited both PGF2alpha- and lipopolysaccharide-induced preterm labor. PDC113.824 inhibited PGF2alpha-mediated, G(alpha)(12)-dependent activation of the Rho/ROCK signaling pathways, actin remodeling, and contraction of human myometrial cells likely by acting as a non-competitive, allosteric modulator of PGF2alpha binding. In contrast to its negative allosteric modulating effects on Rho/ROCK signaling, PDC113.824 acted as a positive allosteric modulator on PGF2alpha-mediated protein kinase C and ERK1/2 signaling. This bias in receptor-dependent signaling was explained by an increase in FP receptor coupling to G(alpha)(q), at the expense of coupling to G(alpha)(12). Our findings regarding the allosteric and biased nature of PDC113.824 offer new mechanistic insights into FP receptor signaling relevant to parturition and suggest novel therapeutic opportunities for the development of new tocolytic drugs.


Assuntos
Dinoprosta/metabolismo , Parto/efeitos dos fármacos , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Quinases Associadas a rho/metabolismo , Regulação Alostérica/efeitos dos fármacos , Animais , Linhagem Celular , Feminino , Imunofluorescência , Humanos , Camundongos , Trabalho de Parto Prematuro/induzido quimicamente , Trabalho de Parto Prematuro/tratamento farmacológico , Peptídeos/síntese química , Peptídeos/uso terapêutico , Gravidez , Proteína Quinase C/metabolismo
8.
Am J Physiol Cell Physiol ; 296(2): C285-95, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19052260

RESUMO

The vectorial transport of ions and water across epithelial cells depends to a large extent on the coordination of the apical and basolateral ion fluxes with energy supply. In this work we provide the first evidence for a regulation by the 5'-AMP-activated protein kinase (AMPK) of the calcium-activated potassium channel KCa3.1 expressed at the basolateral membrane of a large variety of epithelial cells. Inside-out patch-clamp experiments performed on human embryonic kidney (HEK) cells stably transfected with KCa3.1 first revealed a decrease in KCa3.1 activity following the internal addition of AMP at a fixed ATP concentration. This effect was dose dependent with half inhibition at 140 muM AMP in 1 mM ATP. Evidence for an interaction between the COOH-terminal region of KCa3.1 and the gamma1-subunit of AMPK was next obtained by two-hybrid screening and pull-down experiments. Our two-hybrid analysis confirmed in addition that the amino acids extending from Asp(380) to Ala(400) in COOH-terminal were essential for the interaction AMPK-gamma1/KCa3.1. Inside-out experiments on cells coexpressing KCa3.1 with the dominant negative AMPK-gamma1-R299G mutant showed a reduced sensitivity of KCa3.1 to AMP, arguing for a functional link between KCa3.1 and the gamma1-subunit of AMPK. More importantly, coimmunoprecipitation experiments carried out on bronchial epithelial NuLi cells provided direct evidence for the formation of a KCa3.1/AMPK-gamma1 complex at endogenous AMPK and KCa3.1 expression levels. Finally, treating NuLi monolayers with the membrane permeant AMPK activator 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) caused a significant decrease of the KCa3.1-mediated short-circuit currents, an effect reversible by coincubation with the AMPK inhibitor Compound C. These observations argue for a regulation of KCa3.1 by AMPK in a functional epithelium through protein/protein interactions involving the gamma1-subunit of AMPK.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Células Epiteliais/enzimologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Ativação do Canal Iônico , Mucosa Respiratória/enzimologia , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Polaridade Celular , Células Cultivadas , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Células Epiteliais/efeitos dos fármacos , Humanos , Imunoprecipitação , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Ativação do Canal Iônico/efeitos dos fármacos , Transporte de Íons , Potenciais da Membrana , Mutação , Técnicas de Patch-Clamp , Ligação Proteica , Proteínas Recombinantes/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Ribonucleotídeos/farmacologia , Transfecção , Técnicas do Sistema de Duplo-Híbrido
9.
Mol Biol Cell ; 28(26): 3789-3800, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29074566

RESUMO

Stable cytoplasmic bridges arise from failed cytokinesis, the last step of cell division, and are a key feature of syncytial architectures in the germline of most metazoans. Whereas the Caenorhabditiselegans germline is syncytial, its formation remains poorly understood. We found that the germline precursor blastomere, P4 , fails cytokinesis, leaving a stable cytoplasmic bridge between the two daughter cells, Z2 and Z3 Depletion of several regulators of actomyosin contractility resulted in a regression of the membrane partition between Z2 and Z3, indicating that they are required to stabilize the cytoplasmic bridge. Epistatic analysis revealed a pathway in which Rho regulators promote accumulation of the noncannonical anillin ANI-2 at the stable cytoplasmic bridge, which in turns promotes the accumulation of the nonmuscle myosin II NMY-2 and the midbody component CYK-7 at the bridge, in part by limiting the accumulation of canonical anillin ANI-1. Our results uncover key steps in C. elegans germline formation and define a set of conserved regulators that are enriched at the primordial germ cell cytoplasmic bridge to ensure its stability during embryonic development.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Citocinese/fisiologia , Proteínas dos Microfilamentos/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Contráteis/metabolismo , Proteínas Contráteis/fisiologia , Citoplasma/metabolismo , Citoplasma/fisiologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Células Germinativas/metabolismo , Células Germinativas/fisiologia , Proteínas dos Microfilamentos/genética , Contração Muscular , Miosina não Muscular Tipo IIA/metabolismo , Interferência de RNA
10.
J Cell Biol ; 206(1): 129-43, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24982432

RESUMO

Cytokinesis generally produces two separate daughter cells, but in some tissues daughter nuclei remain connected to a shared cytoplasm, or syncytium, through incomplete cytokinesis. How syncytia form remains poorly understood. We studied syncytial formation in the Caenorhabditis elegans germline, in which germ cells connect to a shared cytoplasm core (the rachis) via intercellular bridges. We found that syncytial architecture initiates early in larval development, and germ cells become progressively interconnected until adulthood. The short Anillin family scaffold protein ANI-2 is enriched at intercellular bridges from the onset of germ cell specification, and ANI-2 loss resulted in destabilization of intercellular bridges and germ cell multinucleation defects. These defects were partially rescued by depleting the canonical Anillin ANI-1 or blocking cytoplasmic streaming. ANI-2 is also required for elastic deformation of the gonad during ovulation. We propose that ANI-2 promotes germ cell syncytial organization and allows for compensation of the mechanical stress associated with oogenesis by conferring stability and elasticity to germ cell intercellular bridges.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Células Gigantes/fisiologia , Junções Intercelulares/metabolismo , Proteínas dos Microfilamentos/fisiologia , Animais , Caenorhabditis elegans/citologia , Corrente Citoplasmática , Feminino , Células Germinativas/metabolismo , Gônadas/citologia , Larva/citologia , Larva/metabolismo , Masculino , Oogênese , Estabilidade Proteica
11.
Br J Pharmacol ; 168(5): 1101-3, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23121476

RESUMO

UNLABELLED: Dimers of GPCRs have held the imagination of researchers for almost 20 years. However, only recently has their value as potentially novel drug targets been increased significantly, and primarily, in the context of GPCR heterodimers. The view of receptor heterodimers as allosteric machines has transformed the way we understand structural and functional asymmetries inherent in their organization. These asymmetries alter both signalling output and how they might be targeted pharmacologically. The paper in this issue of BJP by Siddiquee and colleagues () highlights our growing understanding of such asymmetries and their implications. They show that heterodimers of the angiotensin II AT1 receptor and the apelin receptor recognize and respond to their respective ligands in distinct ways from the parent receptors expressed alone. Further, they demonstrate asymmetric allosteric effects in the context of the heterodimer that may have significant implications for our understanding of such receptor complexes. LINKED ARTICLE: This article is a commentary on the research paper by Siddiquee et al., pp. 1104-1117 of this issue. To view this paper visit http://dx.doi.org/10.1111/j.1476-5381.2012.02192.x.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Apelina , Humanos
12.
Methods Cell Biol ; 117: 165-79, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143977

RESUMO

G protein-coupled receptors (GPCRs) represent one of the largest families of cell surface receptors as key targets for pharmacological manipulation. G proteins have long been recognized as allosteric modulators of GPCR ligand binding. More recently, small molecule allosteric modulators have now been widely characterized for a number of GPCRs, and some are now used clinically. Many studies have also underscored the importance of GPCR dimerization or higher-order oligomerization in the control of the physiological responses they modulate. Thus, allosterism can also, between monomer equivalents in the context of a dimer, oligomer, or receptor mosaic, influence signaling pathways downstream. It therefore becomes essential to characterize both small molecule allosteric ligands and allosteric interactions between receptors modulated by canonical orthosteric ligands, in a pathway-specific manner. Here, we describe a simple, radioligand-binding method, which is designed to probe for allosteric modulation mediated by any GPCR interactor, from small molecules to interacting proteins. It can also detect allosteric asymmetries within a GPCR heterodimer, via orthosteric or allosteric ligands. This assay measures time-dependent ligand occupancy of radiolabeled orthosteric or (with adaptations) allosteric ligands as modulated by either small molecules or receptor dimer partners bound or unbound with their own ligands.


Assuntos
Dinoprosta/química , Guanosina 5'-O-(3-Tiotrifosfato)/química , Ensaio Radioligante , Receptor Tipo 1 de Angiotensina/química , Receptores de Prostaglandina/química , Regulação Alostérica , Dinoprosta/genética , Dinoprosta/metabolismo , Expressão Gênica , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Cinética , Ligantes , Multimerização Proteica , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Transdução de Sinais , Transfecção , Trítio
13.
Cell Signal ; 24(1): 333-41, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21964067

RESUMO

The Gq-coupled oxytocin receptor (OTR) and the Gs-coupled ß(2)-adrenergic receptor (ß(2)AR) are both expressed in myometrial cells and mediate uterine contraction and relaxation, respectively. The two receptors represent important pharmacological targets as OTR antagonists and ß(2)AR agonists are used to control pre-term uterine contractions. Despite their physiologically antagonistic effects, both receptors activate the MAP kinases ERK1/2, which has been implicated in uterine contraction and the onset of labor. To determine the signalling pathways involved in mediating the ERK1/2 response, we assessed the effect of blockers of specific G protein-associated pathways. In human myometrial hTERT-C3 cells, inhibition of Gαi as well as inhibition of the Gαq/PKC pathway led to a reduction of both OTR- and ß(2)AR-mediated ERK1/2 activation. The involvement of Gαq/PKC in ß(2)AR-mediated ERK1/2 induction was unexpected. To test whether the emergence of this novel signalling mechanism was dependent on OTR expression in the same cell, we conducted experiments in HEK 293 cells that were transfected with the ß(2)AR alone or co-transfected with the OTR. Using this approach, we found that ß(2)AR-mediated ERK1/2 responses became sensitive to PKC inhibition only in cells co-transfected with the OTR. Inhibitor studies indicated the involvement of an atypical PKC isoform in this process. We confirmed the specific involvement of PKCζ in this pathway by assessing PKCζ translocation to the cell membrane. Consistent with our inhibitor studies, we found that ß(2)AR-mediated PKCζ translocation was dependent on co-expression of OTR. The present demonstration of a novel ß(2)AR-coupled signalling pathway that is dependent on OTR co-expression is suggestive of a molecular interaction between the two receptors.


Assuntos
Ativação Enzimática , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miométrio/citologia , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Ocitocina/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Androstadienos/farmacologia , Carbazóis/farmacologia , Linhagem Celular , Receptores ErbB/metabolismo , Feminino , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Humanos , Indóis , Isoproterenol/farmacologia , Cinética , Sistema de Sinalização das MAP Quinases , Maleimidas , Ocitocina/farmacologia , Ocitocina/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Receptores de Ocitocina/agonistas , Proteínas Recombinantes/metabolismo , Wortmanina
14.
Mol Endocrinol ; 26(7): 1189-202, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22638073

RESUMO

The G protein-coupled prostaglandin F2α (PGF2α) receptor [F prostanoid (FP) receptor] has been implicated in many physiological events including cardiovascular, respiratory, immune, reproductive, and endocrine responses. Binding of PGF2α to FP receptor elicits inositol production and protein kinase C-dependent MAPK activation through Gα(q) coupling. Here we report that AL-8810, previously characterized as an orthosteric antagonist of PGF2α-dependent, Gα(q)-mediated signaling, potently activates ERK1/2 in a protein kinase C-independent manner. Rather, AL-8810 promoted ERK1/2 activation via an epidermal growth factor receptor transactivation mechanism in both human embryonic kidney 293 cells and in the MG-63 osteoblast-like cells, which express endogenous FP receptors. Neither AL-8810- nor PGF2α-mediated stimulation of FP receptor promoted association with ß-arrestins, suggesting that MAPK activation induced by these ligands is independent of ß-arrestin's signaling scaffold functions. Interestingly, the spatiotemporal activation of ERK1/2 promoted by AL-8810 and PGF2α showed almost completely opposite responses in the nucleus and the cytosol. Finally, using [(3)H]thymidine incorporation, we noted differential regulation of PGF2α- and AL-8810-induced cell proliferation in MG-63 cells. This study reveals, for the first time, the signaling biased nature of FP receptor orthosteric ligands toward MAPK signaling. Our findings on the specific patterns of ERK1/2 activation promoted by FP receptor ligands may help dissect the distinct roles of MAPK in FP receptor-dependent physiological responses.


Assuntos
Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores de Prostaglandina/metabolismo , Arrestinas/metabolismo , Proliferação de Células , Dinoprosta/análogos & derivados , Dinoprosta/metabolismo , Dinoprosta/farmacologia , Células HEK293 , Humanos , Inositol/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ligação Proteica , Proteína Quinase C/metabolismo , beta-Arrestinas
15.
Cell Signal ; 24(1): 342-50, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21963428

RESUMO

The oxytocin receptor (OTR) and the ß(2)-adrenergic receptor (ß(2)AR) are key regulators of uterine contraction. These two receptors are targets of tocolytic agents used to inhibit pre-term labor. Our recent study on the nature of OTR- and ß(2)AR-mediated ERK1/2 activation in human hTERT-C3 myometrial cells suggested the presence of an OTR/ß(2)AR hetero-oligomeric complex (see companion article). The goal of this study was to investigate potential allosteric interactions between OTR and ß(2)AR and establish the nature of the interactions between these receptors in myometrial cells. We found that OTR-mediated ERK1/2 activation was attenuated significantly when cells were pretreated with the ß(2)AR agonist isoproterenol or two antagonists, propranolol or timolol. In contrast, pretreatment of cells with a third ß(2)AR antagonist, atenolol resulted in an increase in OTR-mediated ERK1/2 activation. Similarly, ß(2)AR-mediated ERK1/2 activation was strongly attenuated by pretreatment with the OTR antagonists, atosiban and OTA. Physical interactions between OTR and ß(2)AR were demonstrated using co-immunoprecipitation, bioluminescence resonance energy transfer (BRET) and protein-fragment complementation (PCA) assays in HEK 293 cells, the latter experiments indicating the interactions between the two receptors were direct. Our analyses suggest physical interactions between OTR and ß(2)AR in the context of a new heterodimer pair lie at the heart of the allosteric effects.


Assuntos
Ativação Enzimática , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Multimerização Proteica , Receptores Adrenérgicos beta 2/metabolismo , Receptores de Ocitocina/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Regulação Alostérica , Atenolol/farmacologia , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Humanos , Imunoprecipitação , Isoproterenol/farmacologia , Sistema de Sinalização das MAP Quinases , Miométrio/citologia , Ocitocina/farmacologia , Ocitocina/fisiologia , Fosforilação , Propranolol/farmacologia , Ligação Proteica , Receptores de Ocitocina/agonistas , Receptores de Ocitocina/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Timolol/farmacologia , Vasotocina/análogos & derivados , Vasotocina/farmacologia
16.
J Med Chem ; 54(17): 6085-97, 2011 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-21774512

RESUMO

The prostaglandin-F2α (PGF2α) receptor (FP) was targeted to develop tocolytic agents for inhibiting preterm labor. Azabicycloalkane and azapeptide mimics 2-10 were synthesized based on the (3S,6S,9S)-indolizidin-2-one amino acid analogue PDC113.824 (1), which was shown to modulate FP by a biased allosteric mechanism, involving both Gαq- and Gα12-mediated signaling pathways, and exhibited significant tocolytic activity delaying preterm labor in a mouse model ( Goupil ; et al. J. Biol. Chem. 2010 , 285 , 25624 - 25636 ). Although changes in azabicycloalkane stereochemistry and ring size caused loss of activity, replacement of the indolizidin-2-one amino acid with azaGly-Pro and azaPhe-Pro gave azapeptides 6 and 8, which reduced PGF2α-induced myometrial contractions, potentiated the effect of PGF2α on Gαq-mediated ERK1/2 activation, and inhibited FP modulation of cell ruffling, a response dependent on the Gα12/RhoA/ROCK signaling pathway. Revealing complementarities of azabicycloalkane and azapeptide mimics, novel probes, and efficient tocolytic agents were made to study allosteric modulation of the FP receptor.


Assuntos
Compostos Aza/farmacologia , Trabalho de Parto Prematuro/prevenção & controle , Fragmentos de Peptídeos/farmacologia , Receptores de Prostaglandina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tocolíticos/farmacologia , Contração Uterina/efeitos dos fármacos , Animais , Compostos Aza/síntese química , Compostos Aza/química , Western Blotting , Células Cultivadas , Dinoprosta/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Recém-Nascido , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Gravidez , Prenhez/efeitos dos fármacos , Tocolíticos/síntese química , Tocolíticos/química , Quinases Associadas a rho/metabolismo
18.
Cell Signal ; 22(8): 1254-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20403427

RESUMO

Following stimulation of G protein-coupled receptors (GPCRs) at the cell surface, heterotrimeric G proteins are activated. Both Galpha and Gbetagamma subunits regulate specific effectors to transmit signals received by the receptor. Recent data suggest potential nuclear localization or translocation of the Gbetagamma subunit. Here, we show that co-expression of the Gbetagamma dimer decreased phorbol 12-myristate 13-acetate (PMA)-stimulated AP-1 gene reporter activity in HEK293 cells as well as the AP-1 dependent gonadotropin-releasing hormone-stimulated human follicle-stimulating hormone beta reporter activity in LbetaT2 gonadotrope cells. Further, we identify Fos transcription factors as novel interactors of the Gbeta1 subunit, using protein fragment complementation assays, as well as co-immunoprecipitation in vivo and in vitro. Fos proteins dimerize with Jun proteins to form activator protein-1 (AP-1) transcription factor complexes, which regulate target gene expression. Gbetagamma did not interfere with the dimerization of Fos and Jun or their ability to bind DNA. Rather, Gbetagamma co-localized with the AP-1 complex in the nucleus and recruited histone deacetylases (HDACs) to inhibit AP-1 transcriptional activity. Our data indicate a novel role for Gbetagamma subunits as transcriptional regulators.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Transcrição Gênica , Animais , Linhagem Celular , Núcleo Celular/química , Subunidades beta da Proteína de Ligação ao GTP/análise , Subunidades gama da Proteína de Ligação ao GTP/análise , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA