Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(24)2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38139265

RESUMO

The cell-surface targeting of neo-synthesized G protein-coupled receptors (GPCRs) involves the recruitment of receptors into COPII vesicles budding at endoplasmic reticulum exit sites (ERESs). This process is regulated for some GPCRs by escort proteins, which facilitate their export, or by gatekeepers that retain the receptors in the ER. PRAF2, an ER-resident four trans- membrane domain protein with cytoplasmic extremities, operates as a gatekeeper for the GB1 protomer of the heterodimeric GABAB receptor, interacting with a tandem di-leucine/RXR retention motif in the carboxyterminal tail of GB1. PRAF2 was also reported to interact in a two-hybrid screen with a peptide corresponding to the carboxyterminal tail of the chemokine receptor CCR5 despite the absence of RXR motifs in its sequence. Using a bioluminescence resonance energy transfer (BRET)-based subcellular localization system, we found that PRAF2 inhibits, in a concentration-dependent manner, the plasma membrane export of CCR5. BRET-based proximity assays and Co-IP experiments demonstrated that PRAF2/CCR5 interaction does not require the presence of a receptor carboxyterminal tail and involves instead the transmembrane domains of both proteins. The mutation of the potential di-leucine/RXR motif contained in the third intracellular loop of CCR5 does not affect PRAF2-mediated retention. It instead impairs the cell-surface export of CCR5 by inhibiting CCR5's interaction with its private escort protein, CD4. PRAF2 and CD4 thus display opposite roles on the cell-surface export of CCR5, with PRAF2 inhibiting and CD4 promoting this process, likely operating at the level of CCR5 recruitment into COPII vesicles, which leave the ER.


Assuntos
Proteínas de Transporte , Proteínas de Membrana , Receptores CCR5 , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Leucina/metabolismo , Proteínas de Membrana/metabolismo , Transporte Proteico , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de GABA-B/metabolismo , Humanos
2.
Cell Mol Life Sci ; 79(10): 530, 2022 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-36167862

RESUMO

The endoplasmic reticulum exit of some polytopic plasma membrane proteins (PMPs) is controlled by arginin-based retention motifs. PRAF2, a gatekeeper which recognizes these motifs, was shown to retain the GABAB-receptor GB1 subunit in the ER. We report that PRAF2 can interact on a stoichiometric basis with both wild type and mutant F508del Cystic Fibrosis (CF) Transmembrane Conductance Regulator (CFTR), preventing the access of newly synthesized cargo to ER exit sites. Because of its lower abundance, compared to wild-type CFTR, CFTR-F508del recruitment into COPII vesicles is suppressed by the ER-resident PRAF2. We also demonstrate that some pharmacological chaperones that efficiently rescue CFTR-F508del loss of function in CF patients target CFTR-F508del retention by PRAF2 operating with various mechanisms. Our findings open new therapeutic perspectives for diseases caused by the impaired cell surface trafficking of mutant PMPs, which contain RXR-based retention motifs that might be recognized by PRAF2.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Fibrose Cística , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Mutação , Ácido gama-Aminobutírico/metabolismo
3.
Front Endocrinol (Lausanne) ; 13: 883568, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35586623

RESUMO

More than 12 years have passed since the seminal observation that meningococcus, a pathogen causing epidemic meningitis in humans, occasionally associated with infectious vasculitis and septic shock, can promote the translocation of ß-arrestins to the cell surface beneath bacterial colonies. The cellular receptor used by the pathogen to induce signalling in host cells and allowing it to open endothelial cell junctions and reach meninges was unknown. The involvement of ß-arrestins, which are scaffolding proteins regulating G protein coupled receptor signalling and function, incited us to specifically investigate this class of receptors. In this perspective article we will summarize the events leading to the discovery that the ß2-adrenergic receptor is the receptor that initiates the signalling cascades induced by meningococcus in host cells. This receptor, however, cannot mediate cell infection on its own. It needs to be pre-associated with an "early" adhesion receptor, CD147, within a hetero-oligomeric complex, stabilized by the cytoskeletal protein α-actinin 4. It then required several years to understand how the pathogen actually activates the signalling receptor. Once bound to the N-terminal glycans of the ß2-adrenergic receptor, meningococcus provides a mechanical stimulation that induces the biased activation of ß-arrestin-mediated signalling pathways. This activating mechanical stimulus can be reproduced in the absence of any pathogen by applying equivalent forces on receptor glycans. Mechanical activation of the ß2-adrenergic receptor might have a physiological role in signalling events promoted in the context of cell-to-cell interaction.


Assuntos
Neisseria meningitidis , Arrestinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Neisseria meningitidis/metabolismo , Polissacarídeos , beta-Arrestinas/metabolismo
5.
Oncogene ; 40(12): 2243-2257, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33649538

RESUMO

Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein ß-arrestin2 (ß-arr2) regulates tumor suppressor p53 by counteracting Mdm2. ß-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. ß-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. ß-arr2 can be SUMOylated, but no information is available on how SUMO may regulate ß-arr2 nucleocytoplasmic shuttling. While we found ß-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and ß-arr2, via a SUMO interaction motif (SIM), that is required for ß-arr2 cytonuclear trafficking. This SIM promotes association of ß-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective ß-arr2 nuclear entry. Mutation of the SIM inhibits ß-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a ß-arr2 SIM nuclear entry checkpoint, coupled with active ß-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.


Assuntos
Proteínas Ativadoras de GTPase/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína SUMO-1/genética , Proteína Supressora de Tumor p53/genética , beta-Arrestina 2/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Sinais de Exportação Nuclear/genética , Transdução de Sinais/genética , Sumoilação/genética
6.
ACS Pharmacol Transl Sci ; 3(2): 171-178, 2020 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-32296760

RESUMO

Cells are sensitive to chemical stimulation which is converted into intracellular biochemical signals by the activation of specific receptors. Mechanical stimulations can also induce biochemical responses via the activation of various mechano-sensors. Although principally appreciated for their chemosensory function, G-protein-coupled receptors (GPCRs) may participate in mechano-transduction. They are indirectly activated by the paracrine release of chemical compounds secreted in response to mechanical stimuli, but they might additionally behave as mechano-sensors that are directly stimulated by mechanical forces. Although several studies are consistent with this latter hypothesis, the molecular mechanisms of a potential direct mechanical activation of GPCRs have remained elusive until recently. In particular, investigating the activation of the catecholamine ß2-adrenergic receptor by a pathogen revealed that traction forces directly exerted on the N-terminus of the receptor via N-glycan chains activate specific signaling pathways. These findings open new perspectives in GPCR biology and pharmacology since most GPCRs express N-glycan chains in their N-terminus, which might similarly be involved in the interaction with cell-surface glycan-specific lectins in the context of cell-to-cell mechanical signaling.

7.
Cell Mol Life Sci ; 77(24): 5259-5279, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32040695

RESUMO

Focal adhesion kinase (FAK) regulates key biological processes downstream of G protein-coupled receptors (GPCRs) in normal and cancer cells, but the modes of kinase activation by these receptors remain unclear. We report that after GPCR stimulation, FAK activation is controlled by a sequence of events depending on the scaffolding proteins ß-arrestins and G proteins. Depletion of ß-arrestins results in a marked increase in FAK autophosphorylation and focal adhesion number. We demonstrate that ß-arrestins interact directly with FAK and inhibit its autophosphorylation in resting cells. Both FAK-ß-arrestin interaction and FAK inhibition require the FERM domain of FAK. Following the stimulation of the angiotensin receptor AT1AR and subsequent translocation of the FAK-ß-arrestin complex to the plasma membrane, ß-arrestin interaction with the adaptor AP-2 releases inactive FAK from the inhibitory complex, allowing its activation by receptor-stimulated G proteins and activation of downstream FAK effectors. Release and activation of FAK in response to angiotensin are prevented by an AP-2-binding deficient ß-arrestin and by a specific inhibitor of ß-arrestin/AP-2 interaction; this inhibitor also prevents FAK activation in response to vasopressin. This previously unrecognized mechanism of FAK regulation involving a dual role of ß-arrestins, which inhibit FAK in resting cells while driving its activation at the plasma membrane by GPCR-stimulated G proteins, opens new potential therapeutic perspectives in cancers with up-regulated FAK.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/genética , Complexos Multiproteicos/genética , Neoplasias/genética , beta-Arrestinas/genética , Complexo 2 de Proteínas Adaptadoras/genética , Animais , Membrana Celular/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas de Ligação ao GTP/genética , Células HEK293 , Humanos , Camundongos , Complexos Multiproteicos/metabolismo , Neoplasias/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Ligação Proteica/genética , Domínios Proteicos/genética , Receptor Tipo 1 de Angiotensina/genética , Receptores Acoplados a Proteínas G/genética , Vasopressinas/farmacologia
8.
Methods Mol Biol ; 1957: 139-158, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30919352

RESUMO

ß-Arrestins 1 and 2 (ß-arr1 and ß-arr2) are ubiquitous proteins with common and distinct functions. They were initially identified as proteins recruited to stimulated G protein-coupled receptors (GPCRs), regulating their desensitization and internalization. The discovery that ß-arrs could also interact with more than 400 non-GPCR protein partners brought to light their central roles as multifunctional scaffold proteins regulating multiple signalling pathways from the plasma membrane to the nucleus, downstream of GPCRs or independently from these receptors. Through the regulation of the activities and subcellular localization of their binding partners, ß-arrs control various cell processes such as proliferation, cytoskeletal rearrangement, cell motility, and apoptosis. Thus, the identification of ß-arrs binding partners and the characterization of their mode of interaction in cells are central to the understanding of their function. Here we provide methods to explore the molecular interaction of ß-arrs with other proteins in cellulo.


Assuntos
Mapeamento de Interação de Proteínas/métodos , beta-Arrestinas/metabolismo , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Células HEK293 , Humanos , Imunoprecipitação , Ligação Proteica , Saccharomyces cerevisiae/metabolismo , Técnicas do Sistema de Duplo-Híbrido
9.
Proc Natl Acad Sci U S A ; 112(37): E5160-8, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26324936

RESUMO

MAPKs are activated in response to G protein-coupled receptor (GPCR) stimulation and play essential roles in regulating cellular processes downstream of these receptors. However, very little is known about the reciprocal effect of MAPK activation on GPCRs. To investigate possible crosstalk between the MAPK and GPCRs, we assessed the effect of ERK1/2 on the activity of several GPCR family members. We found that ERK1/2 activation leads to a reduction in the steady-state cell-surface expression of many GPCRs because of their intracellular sequestration. This subcellular redistribution resulted in a global dampening of cell responsiveness, as illustrated by reduced ligand-mediated G-protein activation and second-messenger generation as well as blunted GPCR kinases and ß-arrestin recruitment. This ERK1/2-mediated regulatory process was observed for GPCRs that can interact with ß-arrestins, such as type-2 vasopressin, type-1 angiotensin, and CXC type-4 chemokine receptors, but not for the prostaglandin F receptor that cannot interact with ß-arrestin, implicating this scaffolding protein in the receptor's subcellular redistribution. Complementation experiments in mouse embryonic fibroblasts lacking ß-arrestins combined with in vitro kinase assays revealed that ß-arrestin-2 phosphorylation on Ser14 and Thr276 is essential for the ERK1/2-promoted GPCR sequestration. This previously unidentified regulatory mechanism was observed after constitutive activation as well as after receptor tyrosine kinase- or GPCR-mediated activation of ERK1/2, suggesting that it is a central node in the tonic regulation of cell responsiveness to GPCR stimulation, acting both as an effector and a negative regulator.


Assuntos
Arrestinas/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Membrana Celular/metabolismo , Citoplasma/metabolismo , Ativação Enzimática , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Ligantes , Camundongos , Dados de Sequência Molecular , Peptídeos/química , Fosforilação , Ligação Proteica , Receptores de Prostaglandina/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , beta-Arrestina 2 , beta-Arrestinas
10.
Nat Commun ; 5: 4431, 2014 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-25028204

RESUMO

Tumour suppressor PTEN is a phosphatase that negatively regulates the PI3K/AKT pathway. The ability to directly monitor PTEN conformation and function in a rapid, sensitive manner is a key step towards developing anti-cancer drugs aimed at enhancing or restoring PTEN-dependent pathways. Here we developed an intramolecular bioluminescence resonance energy transfer (BRET)-based biosensor, capable of detecting signal-dependent PTEN conformational changes in live cells. The biosensor retains intrinsic properties of PTEN, enabling structure-function and kinetic analyses. BRET shifts, indicating conformational change, were detected following mutations that disrupt intramolecular PTEN interactions, promoting plasma membrane targeting and also following physiological PTEN activation. Using the biosensor as a reporter, we uncovered PTEN activation by several G protein-coupled receptors, previously unknown as PTEN regulators. Trastuzumab, used to treat ERBB2-overexpressing breast cancers also elicited activation-associated PTEN conformational rearrangement. We propose the biosensor can be used to identify pathways regulating PTEN or molecules that enhance its anti-tumour activity.


Assuntos
Técnicas Biossensoriais/métodos , PTEN Fosfo-Hidrolase/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Immunoblotting , Imunoprecipitação , PTEN Fosfo-Hidrolase/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
11.
Handb Exp Pharmacol ; 219: 405-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24292842

RESUMO

Non-visual arrestins were initially appreciated for the roles they play in the negative regulation of G protein-coupled receptors through the processes of desensitisation and endocytosis. The arrestins are also now known as protein scaffolding platforms that act downstream of multiple types of receptors, ensuring relevant transmission of information for an appropriate cellular response. They function as regulatory hubs in several important signalling pathways that are often dysregulated in human cancers. Interestingly, several recent studies have documented changes in expression and localisation of arrestins that occur during cancer progression and that correlate with clinical outcome. Here, we discuss these advances and how changes in expression/localisation may affect functional outputs of arrestins in cancer biology.


Assuntos
Arrestinas/metabolismo , Neoplasias/patologia , Transdução de Sinais , Animais , Humanos , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas
12.
EMBO J ; 30(13): 2557-68, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21642958

RESUMO

The tumour suppressor PTEN (phosphatase and tensin deleted on chromosome 10) regulates major cellular functions via lipid phosphatase-dependent and -independent mechanisms. Despite its fundamental pathophysiological importance, how PTEN's cellular activity is regulated has only been partially elucidated. We report that the scaffolding proteins ß-arrestins (ß-arrs) are important regulators of PTEN. Downstream of receptor-activated RhoA/ROCK signalling, ß-arrs activate the lipid phosphatase activity of PTEN to negatively regulate Akt and cell proliferation. In contrast, following wound-induced RhoA activation, ß-arrs inhibit the lipid phosphatase-independent anti-migratory effects of PTEN. ß-arrs can thus differentially control distinct functional outputs of PTEN important for cell proliferation and migration.


Assuntos
Arrestinas/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/fisiologia , Animais , Arrestinas/antagonistas & inibidores , Arrestinas/genética , Arrestinas/fisiologia , Células COS , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , PTEN Fosfo-Hidrolase/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , beta-Arrestinas
13.
Cell ; 143(7): 1149-60, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-21183077

RESUMO

Following pilus-mediated adhesion to human brain endothelial cells, meningococcus (N. meningitidis), the bacterium causing cerebrospinal meningitis, initiates signaling cascades, which eventually result in the opening of intercellular junctions, allowing meningeal colonization. The signaling receptor activated by the pathogen remained unknown. We report that N. meningitidis specifically stimulates a biased ß2-adrenoceptor/ß-arrestin signaling pathway in endothelial cells, which ultimately traps ß-arrestin-interacting partners, such as the Src tyrosine kinase and junctional proteins, under bacterial colonies. Cytoskeletal reorganization mediated by ß-arrestin-activated Src stabilizes bacterial adhesion to endothelial cells, whereas ß-arrestin-dependent delocalization of junctional proteins results in anatomical gaps used by bacteria to penetrate into tissues. Activation of ß-adrenoceptor endocytosis with specific agonists prevents signaling events downstream of N. meningitidis adhesion and inhibits bacterial crossing of the endothelial barrier. The identification of the mechanism used for hijacking host cell signaling machineries opens perspectives for treatment and prevention of meningococcal infection.


Assuntos
Arrestinas/metabolismo , Encéfalo/microbiologia , Células Endoteliais/microbiologia , Infecções Meningocócicas/metabolismo , Neisseria meningitidis/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Aderência Bacteriana , Barreira Hematoencefálica , Linhagem Celular , Humanos , Infecções Meningocócicas/microbiologia , beta-Arrestinas
14.
J Neurosci ; 30(23): 7804-16, 2010 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-20534829

RESUMO

c-Jun N-terminal kinases (JNKs) (comprising JNK1-3 isoforms) are members of the MAPK (mitogen-activated protein kinase) family, activated in response to various stimuli including growth factors and inflammatory cytokines. Their activation is facilitated by scaffold proteins, notably JNK-interacting protein-1 (JIP1). Originally considered to be mediators of neuronal degeneration in response to stress and injury, recent studies support a role of JNKs in early stages of neurite outgrowth, including adult axonal regeneration. However, the function of individual JNK isoforms, and their potential effector molecules, remained unknown. Here, we analyzed the role of JNK signaling during axonal regeneration from adult mouse dorsal root ganglion (DRG) neurons, combining pharmacological JNK inhibition and mice deficient for each JNK isoform and for JIP1. We demonstrate that neuritogenesis is delayed by lack of JNK2 and JNK3, but not JNK1. JNK signaling is further required for sustained neurite elongation, as pharmacological JNK inhibition resulted in massive neurite retraction. This function relies on JNK1 and JNK2. Neurite regeneration of jip1(-/-) DRG neurons is affected at both initiation and extension stages. Interestingly, activated JNKs (phospho-JNKs), as well as JIP1, are also present in the cytoplasm of sprouting or regenerating axons, suggesting a local action on cytoskeleton proteins. Indeed, we have shown that JNK1 and JNK2 regulate the phosphorylation state of microtubule-associated protein MAP1B, whose role in axonal regeneration was previously characterized. Moreover, lack of MAP1B prevents neurite retraction induced by JNK inhibition. Thus, signaling by individual JNKs is differentially implicated in the reorganization of the cytoskeleton, and neurite regeneration.


Assuntos
Gânglios Espinais/citologia , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Regeneração Nervosa/fisiologia , Neuritos/fisiologia , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Western Blotting , Células Cultivadas , Feminino , Genótipo , Isoenzimas , Proteínas Quinases JNK Ativadas por Mitógeno/deficiência , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , Reação em Cadeia da Polimerase
15.
J Biol Chem ; 284(50): 34769-76, 2009 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19776009

RESUMO

Focal adhesion kinase (FAK) regulates numerous cellular functions and is critical for processes ranging from embryo development to cancer progression. Although autophosphorylation on Tyr-397 appears required for FAK functions in vitro, its role in vivo has not been established. We addressed this question using a mutant mouse (fakDelta) deleted of exon 15, which encodes Tyr-397. The resulting mutant protein FAKDelta is an active kinase expressed at normal levels. Our results demonstrate that the requirement for FAK autophosphorylation varies during development. FAK(Delta/Delta) embryos developed normally up to embryonic day (E) 12.5, contrasting with the lethality at E8.5 of FAK-null embryos. Thus, autophosphorylation on Tyr-397 is not required for FAK to achieve its functions until late mid-gestation. However, FAK(Delta/Delta) embryos displayed hemorrhages, edema, delayed artery formation, vascular remodeling defects, multiple organ abnormalities, and overall developmental retardation at E13.5-14.5, and died thereafter demonstrating that FAK autophosphorylation is also necessary for normal development. Fibroblasts derived from mutant embryos had a normal stellate morphology and expression of focal adhesion proteins, Src family members, p53, and Pyk2. In contrast, in FAK(Delta/Delta) fibroblasts and endothelial cells, spreading and lamellipodia formation were altered with an increased size and number of focal adhesions, enriched in FAKDelta. FAK mutation also decreased fibroblast proliferation. These results show that the physiological functions of FAK in vivo are achieved through both autophosphorylation-independent and autophosphorylation-dependent mechanisms.


Assuntos
Embrião de Mamíferos/enzimologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Mutação , Animais , Biomarcadores/metabolismo , Adesão Celular/fisiologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/anatomia & histologia , Proteína-Tirosina Quinases de Adesão Focal/genética , Integrinas/genética , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação
16.
Mol Cell Neurosci ; 42(4): 288-95, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19683575

RESUMO

We characterized the interactions between plasminogen and neurons and investigated the associated effects on extracellular matrix proteolysis, cell morphology, adhesion, signaling and survival. Upon binding of plasminogen to neurons, the plasmin formed by constitutively expressed tissue plasminogen activator (tPA) degrades extracellular matrix proteins, leading to retraction of the neuron monolayer that detaches from the matrix. This sequence of events required both interaction of plasminogen with carboxy-terminal lysine residues and the proteolytic activity of plasmin. Surprisingly, 24h after plasminogen addition, plasmin-detached neurons survived and remained associated in clusters maintaining focal adhesion kinase phosphorylation contrasting with other adherent cell types fully dissociated by plasmin. However, long-term incubation (72 h) with plasminogen was associated with an increased rate of apoptosis, suggesting that prolonged exposure to plasmin may cause neurotoxicity. Regulation of neuronal organization and survival by plasminogen may be of pathophysiological relevance, as plasminogen is expressed in the brain and/or extravasate during vascular accidents or inflammatory processes.


Assuntos
Sobrevivência Celular/fisiologia , Neurônios/fisiologia , Plasminogênio/metabolismo , Ácido Aminocaproico/metabolismo , Animais , Antifibrinolíticos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Ativação Enzimática , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tecidual/metabolismo
17.
Nature ; 453(7197): 879-84, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18496528

RESUMO

Dopamine orchestrates motor behaviour and reward-driven learning. Perturbations of dopamine signalling have been implicated in several neurological and psychiatric disorders, and in drug addiction. The actions of dopamine are mediated in part by the regulation of gene expression in the striatum, through mechanisms that are not fully understood. Here we show that drugs of abuse, as well as food reinforcement learning, promote the nuclear accumulation of 32-kDa dopamine-regulated and cyclic-AMP-regulated phosphoprotein (DARPP-32). This accumulation is mediated through a signalling cascade involving dopamine D1 receptors, cAMP-dependent activation of protein phosphatase-2A, dephosphorylation of DARPP-32 at Ser 97 and inhibition of its nuclear export. The nuclear accumulation of DARPP-32, a potent inhibitor of protein phosphatase-1, increases the phosphorylation of histone H3, an important component of nucleosomal response. Mutation of Ser 97 profoundly alters behavioural effects of drugs of abuse and decreases motivation for food, underlining the functional importance of this signalling cascade.


Assuntos
Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Nucleossomos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Recompensa , Transdução de Sinais , Animais , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Dopamina/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/química , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Alimentos , Histonas/metabolismo , Aprendizagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Motivação , Atividade Motora/fisiologia , Neostriado/citologia , Neurônios/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Transporte Proteico , Ratos , Transdução de Sinais/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias
18.
Mol Cell Biol ; 28(7): 2314-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18212043

RESUMO

The chemotropic guidance cue netrin-1 promotes neurite outgrowth through its receptor Deleted in Colorectal Cancer (DCC) via activation of Rac1. The guanine nucleotide exchange factor (GEF) linking netrin-1/DCC to Rac1 activation has not yet been identified. Here, we show that the RhoGEF Trio mediates Rac1 activation in netrin-1 signaling. We found that Trio interacts with the netrin-1 receptor DCC in mouse embryonic brains and that netrin-1-induced Rac1 activation in brain is impaired in the absence of Trio. Trio(-/-) cortical neurons fail to extend neurites in response to netrin-1, while they are able to respond to glutamate. Accordingly, netrin-1-induced commissural axon outgrowth is reduced in Trio(-/-) spinal cord explants, and the guidance of commissural axons toward the floor plate is affected by the absence of Trio. The anterior commissure is absent in Trio-null embryos, and netrin-1/DCC-dependent axonal projections that form the internal capsule and the corpus callosum are defective in the mutants. Taken together, these findings establish Trio as a GEF that mediates netrin-1 signaling in axon outgrowth and guidance through its ability to activate Rac1.


Assuntos
Cones de Crescimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Fatores de Crescimento Neural/fisiologia , Neuropeptídeos/fisiologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Superfície Celular/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas rac de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Encéfalo/anormalidades , Encéfalo/citologia , Encéfalo/embriologia , Células COS , Linhagem Celular , Células Cultivadas/citologia , Chlorocebus aethiops , Receptor DCC , Ativação Enzimática , Feminino , Cones de Crescimento/ultraestrutura , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Netrina-1 , Proteínas Oncogênicas/fisiologia , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Medula Espinal/citologia , Medula Espinal/embriologia , Quinases Ativadas por p21/fisiologia , Proteínas rac1 de Ligação ao GTP
19.
J Cell Sci ; 119(Pt 22): 4634-43, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17062642

RESUMO

The conversion of prion protein (PrP(C)) to its protease-resistant isoform is involved in the pathogenesis of prion diseases. Although PrP(C) is highly expressed in neurons and other cell types, its physiological function still remains elusive. Here, we describe how we evaluated its expression, subcellular localization and putative function in brain endothelial cells, which constitute the blood-brain barrier. We detected its expression in microvascular endothelium in mouse brain sections and at intercellular junctions of freshly isolated brain microvessels and cultured brain endothelial cells of mouse, rat and human origin. PrP(C) co-localized with the adhesion molecule platelet endothelial cell adhesion molecule-1 (PECAM-1); moreover, both PrP(C) and PECAM-1 were present in raft membrane microdomains. Using mixed cultures of wild-type and PrP(C)-deficient mouse brain endothelial cells, we observed that PrP(C) accumulation at cell-cell contacts was probably dependent on homophilic interactions between adjacent cells. Moreover, we report that anti-PrP(C) antibodies unexpectedly inhibited transmigration of U937 human monocytic cells as well as freshly isolated monocytes through human brain endothelial cells. Significant inhibition was observed with various anti-PrP(C) antibodies or blocking anti-PECAM-1 antibodies as control. Our results strongly support the conclusion that PrP(C) is expressed by brain endothelium as a junctional protein that is involved in the trans-endothelial migration of monocytes.


Assuntos
Encéfalo/irrigação sanguínea , Células Endoteliais/fisiologia , Junções Intercelulares/metabolismo , Monócitos/fisiologia , Príons/metabolismo , Animais , Movimento Celular , Células Cultivadas , Humanos , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/citologia , Microcirculação/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Príons/genética , Transporte Proteico , Ratos
20.
BMC Genomics ; 7: 198, 2006 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-16889663

RESUMO

BACKGROUND: Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase critical for processes ranging from embryo development to cancer progression. Although isoforms with specific molecular and functional properties have been characterized in rodents and chicken, the organization of FAK gene throughout phylogeny and its potential to generate multiple isoforms are not well understood. Here, we study the phylogeny of FAK, the organization of its gene, and its post-transcriptional processing in rodents and human. RESULTS: A single orthologue of FAK and the related PYK2 was found in non-vertebrate species. Gene duplication probably occurred in deuterostomes after the echinoderma embranchment, leading to the evolution of PYK2 with distinct properties. The amino acid sequence of FAK and PYK2 is conserved in their functional domains but not in their linker regions, with the absence of autophosphorylation site in C. elegans. Comparison of mouse and human FAK genes revealed the existence of multiple combinations of conserved and non-conserved 5'-untranslated exons in FAK transcripts suggesting a complex regulation of their expression. Four alternatively spliced coding exons (13, 14, 16, and 31), previously described in rodents, are highly conserved in vertebrates. Cis-regulatory elements known to regulate alternative splicing were found in conserved alternative exons of FAK or in the flanking introns. In contrast, other reported human variant exons were restricted to Homo sapiens, and, in some cases, other primates. Several of these non-conserved exons may correspond to transposable elements. The inclusion of conserved alternative exons was examined by RT-PCR in mouse and human brain during development. Inclusion of exons 14 and 16 peaked at the end of embryonic life, whereas inclusion of exon 13 increased steadily until adulthood. Study of various tissues showed that inclusion of these exons also occurred, independently from each other, in a tissue-specific fashion. CONCLUSION: The alternative coding exons 13, 14, 16, and 31 are highly conserved in vertebrates and their inclusion in mRNA is tightly but independently regulated. These exons may therefore be crucial for FAK function in specific tissues or during development. Conversely pathological disturbance of the expression of FAK and of its isoforms could lead to abnormal cellular regulation.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/genética , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Processamento Pós-Transcricional do RNA , Regiões 5' não Traduzidas/genética , Processamento Alternativo/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Células Cultivadas , Evolução Molecular , Estruturas Genéticas , Variação Genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Filogenia , Regiões Promotoras Genéticas , Proteínas Tirosina Quinases/genética , Ratos , Elementos Reguladores de Transcrição , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...