Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 8(90): eadd5724, 2023 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-38134242

RESUMO

Tissue-resident CD8+ T cells (TRM) continuously scan peptide-MHC (pMHC) complexes in their organ of residence to intercept microbial invaders. Recent data showed that TRM lodged in exocrine glands scan tissue in the absence of any chemoattractant or adhesion receptor signaling, thus bypassing the requirement for canonical migration-promoting factors. The signals eliciting this noncanonical motility and its relevance for organ surveillance have remained unknown. Using mouse models of viral infections, we report that exocrine gland TRM autonomously generated front-to-back F-actin flow for locomotion, accompanied by high cortical actomyosin contractility, and leading-edge bleb formation. The distinctive mode of exocrine gland TRM locomotion was triggered by sensing physical confinement and was closely correlated with nuclear deformation, which acts as a mechanosensor via an arachidonic acid and Ca2+ signaling pathway. By contrast, naïve CD8+ T cells or TRM surveilling microbe-exposed epithelial barriers did not show mechanosensing capacity. Inhibition of nuclear mechanosensing disrupted exocrine gland TRM scanning and impaired their ability to intercept target cells. These findings indicate that confinement is sufficient to elicit autonomous T cell surveillance in glands with restricted chemokine expression and constitutes a scanning strategy that complements chemosensing-dependent migration.


Assuntos
Linfócitos T CD8-Positivos , Viroses , Camundongos , Animais , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Glândulas Exócrinas , Transdução de Sinais
2.
Front Immunol ; 13: 913366, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35769489

RESUMO

Chemokine-guided leukocyte migration is a hallmark of the immune system to cope with invading pathogens. Intruder confronted dendritic cells (DCs) induce the expression of the chemokine receptor CCR7, which enables them to sense and migrate along chemokine gradients to home to draining lymph nodes, where they launch an adaptive immune response. Chemokine-mediated DC migration is recapitulated and intensively studied in 3D matrix migration chambers. A major caveat in the field is that chemokine gradient formation and maintenance in such 3D environments is generally not assessed. Instead, fluorescent probes, mostly labelled dextran, are used as surrogate molecules, thereby neglecting important electrochemical properties of the chemokines. Here, we used site-specifically, fluorescently labelled CCL19 and CCL21 to study the establishment and shape of the chemokine gradients over time in the 3D collagen matrix. We demonstrate that CCL19 and particularly CCL21 establish stable, but short-distance spanning gradients with an exponential decay-like shape. By contrast, dextran with its neutral surface charge forms a nearly linear gradient across the entire matrix. We show that the charged C-terminal tail of CCL21, known to interact with extracellular matrix proteins, is determinant for shaping the chemokine gradient. Importantly, DCs sense differences in the shape of CCL19 and CCL21 gradients, resulting in distinct spatial migratory responses.


Assuntos
Células Dendríticas , Dextranos , Movimento Celular , Quimiocinas/metabolismo , Dextranos/metabolismo , Receptores CCR7/metabolismo
3.
Front Immunol ; 12: 702453, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603281

RESUMO

Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling.


Assuntos
Linhagem Celular Tumoral , Movimento Celular/imunologia , Células Dendríticas/fisiologia , Receptores CCR7/metabolismo , Humanos
4.
Blood Adv ; 5(1): 99-112, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33570638

RESUMO

The release of newly selected αßT cells from the thymus is key in establishing a functional adaptive immune system. Emigration of the first cohorts of αßT cells produced during the neonatal period is of particular importance, because it initiates formation of the peripheral αßT-cell pool and provides immune protection early in life. Despite this, the cellular and molecular mechanisms of thymus emigration are poorly understood. We examined the involvement of diverse stromal subsets and individual chemokine ligands in this process. First, we demonstrated functional dichotomy in the requirement for CCR7 ligands and identified CCL21, but not CCL19, as an important regulator of neonatal thymus emigration. To explain this ligand-specific requirement, we examined sites of CCL21 production and action and found Ccl21 gene expression and CCL21 protein distribution occurred within anatomically distinct thymic areas. Although Ccl21 transcription was limited to subsets of medullary epithelium, CCL21 protein was captured by mesenchymal stroma consisting of integrin α7+ pericytes and CD34+ adventitial cells at sites of thymic exit. This chemokine compartmentalization involved the heparan sulfate-dependent presentation of CCL21 via its C-terminal extension, explaining the absence of a requirement for CCL19, which lacks this domain and failed to be captured by thymic stroma. Collectively, we identified an important role for CCL21 in neonatal thymus emigration, revealing the importance of this chemokine in initial formation of the peripheral immune system. Moreover, we identified an intrathymic mechanism involving cell-specific production and presentation of CCL21, which demonstrated a functional synergy between thymic epithelial and mesenchymal cells for αßT-cell emigration.


Assuntos
Emigração e Imigração , Linfócitos T , Animais , Animais Recém-Nascidos , Camundongos , Receptores CCR7/genética , Células Estromais
5.
J Leukoc Biol ; 107(6): 1137-1154, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32533638

RESUMO

The chemokine CCL20 is broadly produced by endothelial cells in the liver, the lung, in lymph nodes and mucosal lymphoid tissues, and recruits CCR6 expressing leukocytes, particularly dendritic cells, mature B cells, and subpopulations of T cells. How CCL20 is systemically scavenged is currently unknown. Here, we identify that fluorescently labeled human and mouse CCL20 are efficiently taken-up by the atypical chemokine receptor ACKR4. CCL20 shares ACKR4 with the homeostatic chemokines CCL19, CCL21, and CCL25, although with a lower affinity. We demonstrate that all 4 human chemokines recruit ß-arrestin1 and ß-arrestin2 to human ACKR4. Similarly, mouse CCL19, CCL21, and CCL25 equally activate the human receptor. Interestingly, at the same chemokine concentration, mouse CCL20 did not recruit ß-arrestins to human ACKR4. Further cross-species analysis suggests that human ACKR4 preferentially takes-up human CCL20, whereas mouse ACKR4 similarly internalizes mouse and human CCL20. Furthermore, we engineered a fluorescently labeled chimeric chemokine consisting of the N-terminus of mouse CCL25 and the body of mouse CCL19, termed CCL25_19, which interacts with and is taken-up by human and mouse ACKR4.


Assuntos
Quimiocina CCL19/metabolismo , Quimiocina CCL20/metabolismo , Quimiocina CCL21/metabolismo , Quimiocinas CC/metabolismo , Receptores CCR/metabolismo , beta-Arrestinas/genética , Sequência de Aminoácidos , Animais , Linfócitos B/citologia , Linfócitos B/metabolismo , Sítios de Ligação , Linhagem Celular , Quimiocina CCL19/química , Quimiocina CCL19/genética , Quimiocina CCL20/química , Quimiocina CCL20/genética , Quimiocina CCL21/química , Quimiocina CCL21/genética , Quimiocinas CC/química , Quimiocinas CC/genética , Células HEK293 , Células HeLa , Humanos , Ligantes , Camundongos , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Receptores CCR/química , Receptores CCR/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Transfecção , beta-Arrestinas/metabolismo
6.
Cell Rep ; 29(4): 995-1009.e6, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644919

RESUMO

Chemokine-guided cell migration is pivotal for many immunological and developmental processes. How chemokine receptor signaling persists to guarantee sustained directional migration despite receptor desensitization and internalization remains poorly understood. Here, we uncover a function for an intracellular pool of the chemokine receptor CCR7 present in human dendritic cells and cellular model systems. We find that CCR7 signaling, initiated at the plasma membrane, is translocated by joint trafficking of ß-arrestin and Src kinase to endomembrane-residing CCR7. There, Src tyrosine phosphorylates CCR7, required for the recruitment of Vav1 to form an endomembrane-residing multi-protein signaling complex comprising CCR7, the RhoGEF Vav1, and its effector, Rac1. Interfering with vesicular trafficking affects CCR7-driven cell migration, whereas CCR7:Vav1 interaction at endomembranes is essential for local Rac1 recruitment to CCR7. Photoactivation of Rac1 at endomembranes leads to lamellipodia formation at the cell's leading edge, supporting the role of sustained endomembrane signaling in guiding cell migration.


Assuntos
Membrana Celular/metabolismo , Receptores CCR7/metabolismo , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Movimento Celular , Células Cultivadas , Células HEK293 , Humanos , Camundongos , Ligação Proteica , Proteínas Proto-Oncogênicas c-vav/metabolismo , beta-Arrestinas/metabolismo , Quinases da Família src/metabolismo
7.
Int J Mol Sci ; 20(10)2019 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-31137829

RESUMO

The chemokine receptor CCR7 plays a pivotal role in health and disease. In particular, CCR7 controls homing of antigen-bearing dendritic cells and T cells to lymph nodes, where adaptive immune responses are initiated. However, CCR7 also guides T cells to inflamed synovium and thereby contributes to rheumatoid arthritis and promotes cancer cell migration and metastasis formation. Nanobodies have recently emerged as versatile tools to study G-protein-coupled receptor functions and are being tested in diagnostics and therapeutics. In this study, we designed a strategy to engineer novel nanobodies recognizing human CCR7. We generated a nanobody library based on a solved crystal structure of the nanobody Nb80 recognizing the ß2-adrenergic receptor (ß2AR) and by specifically randomizing two segments within complementarity determining region 1 (CDR1) and CDR3 of Nb80 known to interact with ß2AR. We fused the nanobody library to one half of split-YFP in order to identify individual nanobody clones interacting with CCR7 fused to the other half of split-YFP using bimolecular fluorescence complementation. We present three novel nanobodies, termed Nb1, Nb5, and Nb38, that recognize human CCR7 without interfering with G-protein-coupling and downstream signaling. Moreover, we were able to follow CCR7 trafficking upon CCL19 triggering using Nb1, Nb5, and Nb38.


Assuntos
Receptores CCR7/imunologia , Anticorpos de Domínio Único/imunologia , Afinidade de Anticorpos , Linhagem Celular Tumoral , Células HEK293 , Humanos , Receptores Adrenérgicos beta/imunologia , Receptores CCR7/química , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/imunologia , Anticorpos de Domínio Único/química
8.
Int J Mol Sci ; 19(12)2018 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-30518137

RESUMO

Chemokines are essential guidance cues orchestrating cell migration in health and disease. Cognate chemokine receptors sense chemokine gradients over short distances to coordinate directional cell locomotion. The chemokines CCL19 and CCL21 are essential for recruiting CCR7-expressing dendritic cells bearing pathogen-derived antigens and lymphocytes to lymph nodes, where the two cell types meet to launch an adaptive immune response against the invading pathogen. CCR7-expressing cancer cells are also recruited by CCL19 and CCL21 to metastasize in lymphoid organs. In contrast, atypical chemokine receptors (ACKRs) do not transmit signals required for cell locomotion but scavenge chemokines. ACKR4 is crucial for internalizing and degrading CCL19 and CCL21 to establish local gradients, which are sensed by CCR7-expressing cells. Here, we describe the production of fluorescently tagged chemokines by fusing CCL19 and CCL21 to monomeric red fluorescent protein (mRFP). We show that purified CCL19-mRFP and CCL21-mRFP are versatile and powerful tools to study CCR7 and ACKR4 functions, such as receptor trafficking and chemokine scavenging, in a spatiotemporal fashion. We demonstrate that fluorescently tagged CCL19 and CCL21 permit the visualization and quantification of chemokine gradients in real time, while CCR7-expressing leukocytes and cancer cells sense the guidance cues and migrate along the chemokine gradients.


Assuntos
Quimiocina CCL19/metabolismo , Quimiocina CCL21/metabolismo , Corantes Fluorescentes/metabolismo , Receptores CCR7/metabolismo , Receptores CCR/metabolismo , Animais , Movimento Celular , Colágeno/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células HEK293 , Humanos , Camundongos , Imagem com Lapso de Tempo
9.
Arch Toxicol ; 92(3): 1225-1247, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29164306

RESUMO

Migration of neural crest cells (NCC) is a fundamental developmental process, and test methods to identify interfering toxicants have been developed. By examining cell function endpoints, as in the 'migration-inhibition of NCC (cMINC)' assay, a large number of toxicity mechanisms and protein targets can be covered. However, the key events that lead to the adverse effects of a given chemical or group of related compounds are hard to elucidate. To address this issue, we explored here, whether the establishment of two overlapping structure-activity relationships (SAR)-linking chemical structure on the one hand to a phenotypic test outcome, and on the other hand to a mechanistic endpoint-was useful as strategy to identify relevant toxicity mechanisms. For this purpose, we chose polychlorinated biphenyls (PCB) as a large group of related, but still toxicologically and physicochemically diverse structures. We obtained concentration-dependent data for 26 PCBs in the cMINC assay. Moreover, the test chemicals were evaluated by a new high-content imaging method for their effect on cellular re-distribution of connexin43 and for their capacity to inhibit gap junctions. Non-planar PCBs inhibited NCC migration. The potency (1-10 µM) correlated with the number of ortho-chlorine substituents; non-ortho-chloro (planar) PCBs were non-toxic. The toxicity to NCC partially correlated with gap junction inhibition, while it fully correlated (p < 0.0004) with connexin43 cellular re-distribution. Thus, our double-SAR strategy revealed a mechanistic step tightly linked to NCC toxicity of PCBs. Connexin43 patterns in NCC may be explored as a new endpoint relevant to developmental toxicity screening.


Assuntos
Crista Neural/efeitos dos fármacos , Bifenilos Policlorados/química , Bifenilos Policlorados/toxicidade , Relação Estrutura-Atividade , Animais , Disponibilidade Biológica , Movimento Celular/efeitos dos fármacos , Conexina 43/metabolismo , Junções Comunicantes/efeitos dos fármacos , Humanos , Camundongos , Células NIH 3T3 , Crista Neural/citologia , Bifenilos Policlorados/farmacocinética , Imagem com Lapso de Tempo
10.
FASEB J ; 31(7): 3084-3097, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28360196

RESUMO

The chemokine receptor, CXC chemokine receptor 4 (CXCR4), is selective for CXC chemokine ligand 12 (CXCL12), is broadly expressed in blood and tissue cells, and is essential during embryogenesis and hematopoiesis. CXCL14 is a homeostatic chemokine with unknown receptor selectivity and preferential expression in peripheral tissues. Here, we demonstrate that CXCL14 synergized with CXCL12 in the induction of chemokine responses in primary human lymphoid cells and cell lines that express CXCR4. Combining subactive concentrations of CXCL12 with 100-300 nM CXCL14 resulted in chemotaxis responses that exceeded maximal responses that were obtained with CXCL12 alone. CXCL14 did not activate CXCR4-expressing cells (i.e., failed to trigger chemotaxis and Ca2+ mobilization, as well as signaling via ERK1/2 and the small GTPase Rac1); however, CXCL14 bound to CXCR4 with high affinity, induced redistribution of cell-surface CXCR4, and enhanced HIV-1 infection by >3-fold. We postulate that CXCL14 is a positive allosteric modulator of CXCR4 that enhances the potency of CXCR4 ligands. Our findings provide new insights that will inform the development of novel therapeutics that target CXCR4 in a range of diseases, including cancer, autoimmunity, and HIV.-Collins, P. J., McCully, M. L., Martínez-Muñoz, L., Santiago, C., Wheeldon, J., Caucheteux, S., Thelen, S., Cecchinato, V., Laufer, J. M., Purvanov, V., Monneau, Y. R., Lortat-Jacob, H., Legler, D. F., Uguccioni, M., Thelen, M., Piguet, V., Mellado, M., Moser, B. Epithelial chemokine CXCL14 synergizes with CXCL12 via allosteric modulation of CXCR4.


Assuntos
Quimiocina CXCL12/metabolismo , Quimiocinas CXC/metabolismo , Regulação da Expressão Gênica/fisiologia , Leucócitos Mononucleares/metabolismo , Receptores CXCR4/metabolismo , Sequência de Aminoácidos , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocinas CXC/genética , Quimiotaxia , HIV-1/fisiologia , Humanos , Ligação Proteica , Conformação Proteica , RNA Mensageiro , Receptores CXCR4/genética , Transdução de Sinais
11.
Mol Pharmacol ; 91(4): 331-338, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28082305

RESUMO

Chemokine receptors are seven transmembrane-domain receptors belonging to class A of G-protein-coupled receptors (GPCRs). The receptors together with their chemokine ligands constitute the chemokine system, which is essential for directing cell migration and plays a crucial role in a variety of physiologic and pathologic processes. Given the importance of orchestrating cell migration, it is vital that chemokine receptor signaling is tightly regulated to ensure appropriate responses. Recent studies highlight a key role for cholesterol in modulating chemokine receptor activities. The steroid influences the spatial organization of GPCRs within the membrane bilayer, and consequently can tune chemokine receptor signaling. The effects of cholesterol on the organization and function of chemokine receptors and GPCRs in general include direct and indirect effects (Fig. 1). Here, we review how cholesterol and some key metabolites modulate functions of the chemokine system in multiple ways. We emphasize the role of cholesterol in chemokine receptor oligomerization, thereby promoting the formation of a signaling hub enabling integration of distinct signaling pathways at the receptor-membrane interface. Moreover, we discuss the role of cholesterol in stabilizing particular receptor conformations and its consequence for chemokine binding. Finally, we highlight how cholesterol accumulation, its deprivation, or cholesterol metabolites contribute to modulating cell orchestration during inflammation, induction of an adaptive immune response, as well as to dampening an anti-tumor immune response.


Assuntos
Colesterol/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Membrana Celular/metabolismo , Doença , Humanos , Modelos Biológicos , Receptores de Quimiocinas/química , Transdução de Sinais
12.
Biosci Rep ; 35(6)2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26487707

RESUMO

Drosophila GoLoco motif-containing protein Pins is unusual in its highly efficient interaction with both GDP- and the GTP-loaded forms of the α-subunit of the heterotrimeric Go protein. We analysed the interactions of Gαo in its two nucleotide forms with GoLoco1-the first of the three GoLoco domains of Pins-and the possible structures of the resulting complexes, through combination of conventional fluorescence and FRET measurements as well as through molecular modelling. Our data suggest that the orientation of the GoLoco1 motif on Gαo significantly differs between the two nucleotide states of the latter. In other words, a rotation of the GoLoco1 peptide in respect with Gαo must accompany the nucleotide exchange in Gαo. The sterical hindrance requiring such a rotation probably contributes to the guanine nucleotide exchange inhibitor activity of GoLoco1 and Pins as a whole. Our data have important implications for the mechanisms of Pins regulation in the process of asymmetric cell divisions.


Assuntos
Proteínas de Drosophila/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Inibidores de Dissociação do Nucleotídeo Guanina/genética , Nucleotídeos de Guanina/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Motivos de Aminoácidos/genética , Animais , Divisão Celular Assimétrica/genética , Proteínas de Ciclo Celular , Drosophila/genética , Proteínas de Drosophila/química , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Inibidores de Dissociação do Nucleotídeo Guanina/química , Proteínas Heterotriméricas de Ligação ao GTP/química , Peptídeos/química , Peptídeos/genética
13.
Elife ; 32014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24950964

RESUMO

Homotypic or entotic cell-in-cell invasion is an integrin-independent process observed in carcinoma cells exposed during conditions of low adhesion such as in exudates of malignant disease. Although active cell-in-cell invasion depends on RhoA and actin, the precise mechanism as well as the underlying actin structures and assembly factors driving the process are unknown. Furthermore, whether specific cell surface receptors trigger entotic invasion in a signal-dependent fashion has not been investigated. In this study, we identify the G-protein-coupled LPA receptor 2 (LPAR2) as a signal transducer specifically required for the actively invading cell during entosis. We find that G12/13 and PDZ-RhoGEF are required for entotic invasion, which is driven by blebbing and a uropod-like actin structure at the rear of the invading cell. Finally, we provide evidence for an involvement of the RhoA-regulated formin Dia1 for entosis downstream of LPAR2. Thus, we delineate a signaling process that regulates actin dynamics during cell-in-cell invasion.


Assuntos
Actinas/química , Regulação Neoplásica da Expressão Gênica , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Entose , Forminas , Células HEK293 , Humanos , Invasividade Neoplásica , Neoplasias/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Biochem Pharmacol ; 82(10): 1311-9, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21689640

RESUMO

The Wnt/Frizzled signaling pathway plays multiple functions in animal development and, when deregulated, in human disease. The G-protein coupled receptor (GPCR) Frizzled and its cognate heterotrimeric Gi/o proteins initiate the intracellular signaling cascades resulting in cell fate determination and polarization. In this review, we summarize the knowledge on the ligand recognition, biochemistry, modifications and interacting partners of the Frizzled proteins viewed as GPCRs. We also discuss the effectors of the heterotrimeric Go protein in Frizzled signaling. One group of these effectors is represented by small GTPases of the Rab family, which amplify the initial Wnt/Frizzled signal. Another effector is the negative regulator of Wnt signaling Axin, which becomes deactivated in response to Go action. The discovery of the GPCR properties of Frizzled receptors not only provides mechanistic understanding to their signaling pathways, but also paves new avenues for the drug discovery efforts.


Assuntos
Receptores Frizzled/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Animais , Receptores Frizzled/genética , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Humanos , Proteínas Wnt/genética
15.
Sci Signal ; 3(136): ra65, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20736485

RESUMO

Rab5 is a small guanosine triphosphatase (GTPase) that regulates the early stages of endocytosis and is conserved in eukaryotes. Rab5 regulates the internalization of receptors and other membrane-associated signaling proteins. The function of Rab5 in these processes is considered relatively passive, so that the endocytic capacity of Rab5 is used during, for example, beta-arrestin-dependent internalization of G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs). Direct recruitment or activation of Rab5 by the components of these signaling pathways has not been reported. Here, we demonstrate an interaction of Drosophila Rab5 and an immediate transducer of GPCR signaling, the G protein G(o), in vitro and in vivo. Rab5 and G(o) bound to each other as purified proteins, as well as in fly extracts. In cellular assays, G(o) led to Rab5 activation and endosome fusion. We further showed that the G(o)-Rab5 interaction functioned in Drosophila planar cell polarity and Wingless signal transduction, pathways initiated by GPCRs of the Frizzled (Fz) family. Additionally, the recycling Rab GTPases Rab4 and Rab11 functioned in Fz- and G(o)-mediated signaling to favor planar cell polarity over canonical Wingless signaling. The interplay between heterotrimeric G proteins and Rab GTPases controlled receptor internalization, revealing a previously uncharacterized regulatory mechanism in GPCR signaling.


Assuntos
Endocitose/fisiologia , Endossomos/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Polaridade Celular/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Endossomos/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Ligação Proteica , Receptores Acoplados a Proteínas G/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab4 de Ligação ao GTP/genética , Proteínas rab4 de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/genética
16.
Anal Biochem ; 397(2): 202-7, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19849998

RESUMO

[(35)S]GTPgammaS, the nonhydrolyzable radioactive GTP analog, has been a powerful tool in G protein studies and has set the standards in this field of research. However, its radioactive nature imposes clear limitations to its use in regular laboratory practice and in high-throughput experimentation. The europium-labeled GTP analog (Eu-GTP) has been used as an alternative in the analysis of G protein activation by G protein-coupled receptors in cellular membrane preparations. Here we expand the usage of Eu-GTP and show that it can be applied in other types of assays where [(35)S]GTPgammaS has been previously utilized. We demonstrate the applicability of the modified Eu-GTP binding technology to analysis of heterotrimeric and monomeric G proteins of natural and recombinant sources, from different organisms, in assays with soluble proteins and membrane-containing assays of a high-throughput format. The deci-nanomolar K(D) of Eu-GTP for the tested G proteins is similar to that of other fluorescent-modified GTP analogs, while the sensitivity achieved in time-resolved fluorescence analysis of Eu-GTP exceeds that of the radioactive measurements. Overall, the results of our modified Eu-GTP binding assay present Eu-GTP as a general nonradioactive alternative for G protein studies, especially attractive in high-throughput experiments.


Assuntos
Európio/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/análogos & derivados , Compostos Organometálicos/metabolismo , Animais , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Guanosina Trifosfato/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Cinética , Receptores Acoplados a Proteínas G/metabolismo , Suínos
17.
Curr Microbiol ; 50(4): 217-22, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15902470

RESUMO

Amino acid residues in the active site of quinoline 2-oxidoreductase (Qor) that are deemed important for substrate binding and turnover were replaced by site-directed mutagenesis. The apparent k(cat) values for quinoline were reduced 2.4-, 38-, 40-, and 199-fold in the protein variants QorA259G, QorW331G, QorV373A, and QorA546G, respectively. The substitution A259G did not significantly alter K(m app). Despite the presumed crucial role of W331 and V373 in substrate positioning, the replacements W331G (K(m app): 0.33 mM) and V373A (K(m app): 0.41 mM) only slightly affected affinity for quinoline (K(m app) of Qor: 0.12 mM). QorA546G showed an increased affinity for quinoline and quinoxaline, as suggested by its 4.3- and 7.5-fold decrease in K(m) (app (quinoline))and K(m app (quinoxaline)), respectively, compared with Qor. The relative activities of the protein variants towards substituted quinolines differed from those of Qor. QorW331G, for example, may be suitable for hydroxylation of quinoxaline and C4-substituted quinolines.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Pseudomonas putida/enzimologia , Pseudomonas putida/genética , Especificidade por Substrato/genética
18.
Structure ; 12(8): 1425-35, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15296736

RESUMO

The soil bacterium Pseudomonas putida 86 uses quinoline as a sole source of carbon and energy. Quinoline 2-oxidoreductase (Qor) catalyzes the first metabolic step converting quinoline to 2-oxo-1,2-dihydroquinoline. Qor is a member of the molybdenum hydroxylases. The molybdenum ion is coordinated by two ene-dithiolate sulfur atoms, two oxo-ligands, and a catalytically crucial sulfido-ligand, whose position in the active site was controversial. The 1.8 A resolution crystal structure of Qor indicates that the sulfido-ligand occupies the equatorial position at the molybdenum ion. The structural comparison of Qor with the allopurinol-inhibited xanthine dehydrogenase from Rhodobacter capsulatus allows direct insight into the mechanism of substrate recognition and the identification of putative catalytic residues. The active site protein variants QorE743V and QorE743D were analyzed to assess the catalytic role of E743.


Assuntos
Oxigenases de Função Mista/química , Molibdênio/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Quinolinas/química , Xantina Desidrogenase/química , Alopurinol/química , Sequência de Aminoácidos , Carbono/química , Cristalografia por Raios X , Ligantes , Dados de Sequência Molecular , Pseudomonas putida/enzimologia , Rhodobacter capsulatus/enzimologia , Homologia de Sequência de Aminoácidos , Enxofre/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...