Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cells ; 8(6)2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31216755

RESUMO

First thought to orchestrate exclusively leukocyte trafficking, chemokines are now acknowledged for their multiple roles in the regulation of cell proliferation, differentiation, and survival. Dysregulation of their normal functions contributes to various pathologies, including inflammatory diseases and cancer. The two chemokine receptor 3 variants CXCR3-A and CXCR3-B, together with their cognate chemokines (CXCL11, CXCL10, CXCL9, CXCL4, and CXCL4L1), are involved in the control but also in the development of many tumors. CXCR3-A drives the infiltration of leukocytes to the tumor bed to modulate tumor progression (paracrine axis). Conversely, tumor-driven changes in the expression of the CXCR3 variants and their ligands promote cancer progression (autocrine axis). This review summarizes the anti- and pro-tumoral activities of the CXCR3 variants and their associated chemokines with a focus on the understanding of their distinct biological roles in the tumor microenvironment.


Assuntos
Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Microambiente Tumoral/fisiologia , Quimiocinas/genética , Quimiocinas/metabolismo , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Humanos , Ligantes , Neoplasias/metabolismo , Microambiente Tumoral/genética
2.
Mol Metab ; 12: 76-88, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29735266

RESUMO

OBJECTIVES: Neuraminidase 1 (NEU1) cleaves terminal sialic acids of glycoconjugates during lysosomal catabolism. It also modulates the structure and activity of cellular surface receptors affecting diverse pathways. Previously we demonstrated that NEU1 activates the insulin receptor (IR) and that NEU1-deficient CathAS190A-Neo mice (hypomorph of the NEU1 activator protein, cathepsin A/CathA) on a high-fat diet (HFD) develop hyperglycaemia and insulin resistance faster than wild-type animals. The major objective of the current work was to reveal the molecular mechanism by which NEU1 desialylation activates the IR and to test if increase of NEU1 activity in insulin target tissues reverses insulin resistance and glucose intolerance. METHODS: To test if desialylation causes a conformational change in the IR dimer we measured interaction between the receptor subunits by Bioluminescence Resonance Energy Transfer in the HEK293T cells either overexpressing NEU1 or treated with the NEU1 inhibitor. The influence of NEU1 overexpression on insulin resistance was studied in vitro in palmitate-treated HepG2 cells transduced with NEU1-expressing lentivirus and in vivo in C57Bl6 mice treated with HFD and either pharmacological inducer of NEU1, Ambroxol or NEU1-expressing adenovirus. NEU1-deficient CathAS190A-Neo mice were used as a control. RESULTS: By desialylation of IR, NEU1 induced formation of its active dimer leading to insulin signaling. Overexpression of NEU1 in palmitate-treated HepG2 cells restored insulin signaling, suggesting that increased NEU1 levels may reverse insulin resistance. Five-day treatment of glycemic C57Bl6 mice receiving HFD with the activator of the lysosomal gene network, Ambroxol, increased NEU1 expression and activity in muscle tissue, normalized fasting glucose levels, and improved physiological and molecular responses to glucose and insulin. Ambroxol did not improve insulin sensitivity in obese insulin-resistant CathAS190A-Neo mice indicating that the Ambroxol effect is mediated through NEU1 induction. Sustained increase of liver NEU1 activity through adenovirus-based gene transfer failed to attenuate insulin resistance most probably due to negative feedback regulation of IR expression. CONCLUSION: Together our results demonstrate that increase of NEU1 activity in insulin target tissues reverses insulin resistance and glucose intolerance suggesting that a pharmacological modulation of NEU1 activity may be potentially explored for restoring insulin sensitivity and resolving hyperglycemia associated with T2DM.


Assuntos
Resistência à Insulina , Neuraminidase/metabolismo , Obesidade/metabolismo , Receptor de Insulina/metabolismo , Ambroxol/farmacologia , Ambroxol/uso terapêutico , Animais , Células HEK293 , Células Hep G2 , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Neuraminidase/genética , Obesidade/tratamento farmacológico
3.
Br J Pharmacol ; 175(9): 1419-1438, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29272550

RESUMO

BACKGROUND AND PURPOSE: Chemokines and their receptors form an intricate interaction and signalling network that plays critical roles in various physiological and pathological cellular processes. The high promiscuity and apparent redundancy of this network makes probing individual chemokine/receptor interactions and functional effects, as well as targeting individual receptor axes for therapeutic applications, challenging. Despite poor sequence identity, the N-terminal regions of chemokines, which play a key role in their activity and selectivity, contain several conserved features. Thus far little is known regarding the molecular basis of their interactions with typical and atypical chemokine receptors or the conservation of their contributions across chemokine-receptor pairs. EXPERIMENTAL APPROACH: We used a broad panel of chemokine variants and modified peptides derived from the N-terminal region of chemokines CXCL12, CXCL11 and vCCL2, to compare the contributions of various features to binding and activation of their shared receptors, the two typical, canonical G protein-signalling receptors, CXCR4 and CXCR3, as well as the atypical scavenger receptor CXCR7/ACKR3, which shows exclusively arrestin-dependent activity. KEY RESULTS: We provide molecular insights into the plasticity of the ligand-binding pockets of these receptors, their chemokine binding modes and their activation mechanisms. Although the chemokine N-terminal region is a critical determinant, neither the most proximal residues nor the N-loop are essential for binding and activation of ACKR3, as distinct from binding and activation of CXCR4 and CXCR3. CONCLUSION AND IMPLICATIONS: These results suggest a different interaction mechanism between this atypical receptor and its ligands and illustrate its strong propensity to activation.


Assuntos
Sítios de Ligação , Peptídeos/metabolismo , Ligação Proteica , Receptores CXCR3/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animais , Arrestina/metabolismo , Células Cultivadas , Quimiocina CXCL11/metabolismo , Quimiocina CXCL12/metabolismo , Humanos , Ligantes , Ensaio Radioligante
4.
Sci Signal ; 10(471)2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28325822

RESUMO

Chemokines orchestrate cell migration for development, immune surveillance, and disease by binding to cell surface heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs). The array of interactions between the nearly 50 chemokines and their 20 GPCR targets generates an extensive signaling network to which promiscuity and biased agonism add further complexity. The receptor CXCR4 recognizes both monomeric and dimeric forms of the chemokine CXCL12, which is a distinct example of ligand bias in the chemokine family. We demonstrated that a constitutively monomeric CXCL12 variant reproduced the G protein-dependent and ß-arrestin-dependent responses that are associated with normal CXCR4 signaling and lead to cell migration. In addition, monomeric CXCL12 made specific contacts with CXCR4 that are not present in the structure of the receptor in complex with a dimeric form of CXCL12, a biased agonist that stimulates only G protein-dependent signaling. We produced an experimentally validated model of an agonist-bound chemokine receptor that merged a nuclear magnetic resonance-based structure of monomeric CXCL12 bound to the amino terminus of CXCR4 with a crystal structure of the transmembrane domains of CXCR4. The large CXCL12:CXCR4 protein-protein interface revealed by this structure identified previously uncharacterized functional interactions that fall outside of the classical "two-site model" for chemokine-receptor recognition. Our model suggests a mechanistic hypothesis for how interactions on the extracellular face of the receptor may stimulate the conformational changes required for chemokine receptor-mediated signal transduction.


Assuntos
Quimiocina CXCL12/química , Multimerização Proteica , Receptores CXCR4/química , Transdução de Sinais , Sequência de Aminoácidos , Linhagem Celular Tumoral , Movimento Celular/genética , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células HEK293 , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Mutação , Ligação Proteica , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , beta-Arrestina 2/genética , beta-Arrestina 2/metabolismo
5.
J Med Chem ; 59(16): 7512-24, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27434274

RESUMO

The CXCR4 receptor binds with meaningful affinities only CXCL12 and synthetic antagonists/inverse agonists. We recently described high affinity synthetic agonists for this chemokine receptor, obtained by grafting the CXCL12 N-terminus onto the inverse agonist T140. While those chimeric molecules behave as agonists for CXCR4, their binding and activation mode are unknown. The present SAR of those CXCL12-oligopeptide grafts reveals the key determinants involved in CXCR4 activation. Position 3 (Val) controls affinity, whereas position 7 (Tyr) acts as an efficacy switch. Chimeric molecules bearing aromatic residues in position 3 possess high binding affinities for CXCR4 and are Gαi full agonists with robust chemotactic properties. Fine-tuning of electron-poor aromatic rings in position 7 enhances receptor activation. To rationalize these results, a homology model of a receptor-ligand complex was built using the published crystal structures of CXCR4. Molecular dynamics simulations reveal further details accounting for the observed SAR for this series.


Assuntos
Peptídeos/farmacologia , Receptores CXCR4/agonistas , Relação Dose-Resposta a Droga , Humanos , Simulação de Dinâmica Molecular , Estrutura Molecular , Peptídeos/síntese química , Peptídeos/química , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
6.
Diabetes ; 62(7): 2338-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23520133

RESUMO

Neuraminidases (sialidases) catalyze the removal of sialic acid residues from sialylated glycoconjugates. We now report that mammalian neuraminidase 1 (Neu1), in addition to its catabolic function in lysosomes, is transported to the cell surface where it is involved in the regulation of insulin signaling. Insulin binding to its receptor rapidly induces interaction of the receptor with Neu1, which hydrolyzes sialic acid residues in the glycan chains of the receptor and, consequently, induces its activation. Cells from sialidosis patients with a genetic deficiency of Neu1 show impairment of insulin-induced phosphorylation of downstream protein kinase AKT, and treatment of these cells with purified Neu1 restores signaling. Genetically modified mice with ∼10% of the normal Neu1 activity exposed to a high-fat diet develop hyperglycemia and insulin resistance twice as fast as their wild-type counterparts. Together, these studies identify Neu1 as a novel component of the signaling pathways of energy metabolism and glucose uptake.


Assuntos
Insulina/metabolismo , Mucolipidoses/metabolismo , Neuraminidase/metabolismo , Transdução de Sinais/fisiologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Metabolismo Energético/fisiologia , Fibroblastos/metabolismo , Teste de Tolerância a Glucose , Células HEK293 , Humanos , Insulina/genética , Camundongos , Camundongos Knockout , Mucolipidoses/genética , Neuraminidase/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 304(1): R10-22, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23152113

RESUMO

Acute renal failure (ARF) is a serious medical complication characterized by an abrupt and sustained decline in renal function. Despite significant advances in supportive care, there is currently no effective treatment to restore renal function. PGE(2) is a lipid hormone mediator abundantly produced in the kidney, where it acts locally to regulate renal function; several studies suggest that modulating EP(4) receptor activity could improve renal function following kidney injury. An optimized peptidomimetic ligand of EP(4) receptor, THG213.29, was tested for its efficacy to improve renal function (glomerular filtration rate, renal plasma flow, and urine output) and histological changes in a model of ARF induced by either cisplatin or renal artery occlusion in Sprague-Dawley rats. THG213.29 modulated PGE(2)-binding dissociation kinetics, indicative of an allosteric binding mode. Consistently, THG213.29 antagonized EP(4)-mediated relaxation of piglet saphenous vein rings, partially inhibited EP(4)-mediated cAMP production, but did not affect Gα(i) activation or ß-arrestin recruitment. In vivo, THG213.29 significantly improved renal function and histological changes in cisplatin- and renal artery occlusion-induced ARF models. THG213.29 increased mRNA expression of heme-oxygenase 1, Bcl2, and FGF-2 in renal cortex; correspondingly, in EP(4)-transfected HEK293 cells, THG213.29 augmented FGF-2 and abrogated EP(4)-dependent overexpression of inflammatory IL-6 and of apoptotic death domain-associated protein and BCL2-associated agonist of cell death. Our results demonstrate that THG213.29 represents a novel class of diuretic agent with noncompetitive allosteric modulator effects on EP(4) receptor, resulting in improved renal function and integrity following acute renal failure.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Rim/efeitos dos fármacos , Rim/fisiologia , Oligopeptídeos/uso terapêutico , Receptores de Prostaglandina E Subtipo EP4/agonistas , Recuperação de Função Fisiológica/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Animais , Cisplatino/efeitos adversos , AMP Cíclico/biossíntese , Modelos Animais de Doenças , Cães , Feminino , Fator 2 de Crescimento de Fibroblastos/biossíntese , Taxa de Filtração Glomerular/efeitos dos fármacos , Células HEK293 , Heme Oxigenase-1/biossíntese , Humanos , Interleucina-6/biossíntese , Masculino , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Ratos Sprague-Dawley , Fluxo Plasmático Renal/fisiologia , Veia Safena/efeitos dos fármacos , Veia Safena/patologia , Suínos/fisiologia
8.
Proc Natl Acad Sci U S A ; 108(43): 17655-60, 2011 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-21990345

RESUMO

Chemokines and chemokine receptors are extensively and broadly involved in cancer metastasis. Previously, we demonstrated that epigenetic silencing of the chemokine CXCL12 sensitizes breast and colon cancer cells to endocrine signaling and metastasis to distant tissues. Yet, the precise mechanism whereby CXCL12 production by tumor cells regulates dissemination remains unclear. Here, we show that administration of CXCL12 extended survival of tumor-bearing mice by potently limiting metastasis of colorectal carcinoma or murine melanoma. Because secreted CXCL12 is a mixture of monomeric and dimeric species in equilibrium, oligomeric variants that either promote (monomer) or halt (dimer) chemotaxis were used to dissect the mechanisms interrupting carcinoma metastasis. Monomeric CXCL12 mobilized intracellular calcium, inhibited cAMP signaling, recruited ß-arrestin-2, and stimulated filamentous-actin accumulation and cell migration. Dimeric CXCL12 activated G-protein-dependent calcium flux, adenylyl cyclase inhibition, and the rapid activation of ERK1/2, but only weakly, if at all, recruited arrestin, stimulated actin polymerization, or promoted chemotaxis. NMR analyses illustrated that CXCL12 monomers made specific contacts with CXCR4 that were lost following dimerization. Our results establish the potential for inhibiting CXCR4-mediated metastasis by administration of CXCL12. Chemokine-mediated migration and ß-arrestin responses did not dictate the antitumor effect of CXCL12. We conclude that cellular migration is tightly regulated by selective CXCR4 signaling evoked by unique interactions with distinct ligand quaternary structures.


Assuntos
Quimiocina CXCL12/farmacologia , Neoplasias Colorretais/patologia , Metástase Neoplásica/prevenção & controle , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Actinas/metabolismo , Animais , Arrestinas/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Dimerização , Citometria de Fluxo , Humanos , Camundongos , Ressonância Magnética Nuclear Biomolecular , beta-Arrestina 2 , beta-Arrestinas
9.
Viral Immunol ; 24(1): 11-26, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21319975

RESUMO

Herpes simplex virus type 1 (HSV-1) is a ubiquitously occurring pathogen that infects humans early in childhood. The virus persists as a latent infection in dorsal root ganglia, especially of the trigeminal nerve, and frequently becomes reactivated in humans under conditions of stress. Monocytic cells constitute an important component of the innate and adaptive immune responses. We show here for the first time that HSV-1 stimulates human FasL promoter and induces de novo expression of FasL on the surface of human monocytic cells, including monocytes and macrophages. This virus-induced FasL expression causes death of monocytic cells growing in suspension, but not in monolayers (e.g., macrophages). The addition of a broad-spectrum caspase inhibitor, as well as anti-FasL antibodies, reduced cell death but increased viral replication in the virus-infected cell cultures. We also show here for the first time that the virus-induced de novo expression of FasL on the cell surface acts as an immune evasion mechanism by causing the death of interacting human CD4+ T cells, CD8+ T cells, and natural killer (NK) cells. Our study provides novel insights on FasL expression and cell death in HSV-infected human monocytic cells and their impact on interacting immune cells.


Assuntos
Morte Celular , Proteína Ligante Fas/biossíntese , Expressão Gênica , Herpesvirus Humano 1/patogenicidade , Evasão da Resposta Imune , Monócitos/virologia , Replicação Viral , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/imunologia , Humanos , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Monócitos/imunologia
10.
J Biol Chem ; 285(49): 37939-43, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20956518

RESUMO

CXCR7 is an atypical chemokine receptor that signals through ß-arrestin in response to agonists without detectable activation of heterotrimeric G-proteins. Its cognate chemokine ligand CXCL12 also binds CXCR4, a chemokine receptor of considerable clinical interest. Here we report that TC14012, a peptidomimetic inverse agonist of CXCR4, is an agonist on CXCR7. The potency of ß-arrestin recruitment to CXCR7 by TC14012 is much higher than that of the previously reported CXCR4 antagonist AMD3100 and differs only by one log from that of the natural ligand CXCL12 (EC(50) 350 nM for TC14012, as compared with 30 nM for CXCL12 and 140 µM for AMD3100). Moreover, like CXCL12, TC14012 leads to Erk 1/2 activation in U373 glioma cells that express only CXCR7, but not CXCR4. Given that with TC14012 and AMD3100 two structurally unrelated CXCR4 antagonists turn out to be agonists on CXCR7, this likely reflects differences in the activation mechanism of the arrestin pathway by both receptors. To identify the receptor domain responsible for these opposed effects, we investigated CXCR4 and CXCR7 C terminus-swapping chimeras. Using quantitative bioluminescence resonance energy transfer, we find that the CXCR7 receptor core formed by the seven-transmembrane domains and the connecting loops determines the agonistic activity of both TC14012 and AMD3100. Moreover, we find that the CXCR7 chimera bearing the CXCR4 C-terminal constitutively associates with arrestin in the absence of ligands. Our data suggest that the CXCR4 and CXCR7 cores share ligand-binding surfaces for the binding of the synthetic ligands, indicating that CXCR4 inhibitors should be tested also on CXCR7.


Assuntos
Arrestinas/metabolismo , Oligopeptídeos/farmacologia , Peptidomiméticos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Fármacos Anti-HIV/farmacologia , Arrestinas/genética , Benzilaminas , Linhagem Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ciclamos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Células HEK293 , Compostos Heterocíclicos/farmacologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estrutura Terciária de Proteína , Receptores CXCR/genética , Receptores CXCR4/genética , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , beta-Arrestinas
11.
Biochem Pharmacol ; 78(11): 1382-90, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19631193

RESUMO

CXCR4, a G-protein-coupled receptor, which binds the chemokine stromal cell-derived factor 1 alpha (SDF-1alpha, CXCL12), is one of two co-receptors most frequently used by HIV-1 to infect CD4+ lymphocytes. The SDF-1alpha/CXCR4 axis is also involved in angiogenesis, in stem cell homing to bone marrow, in rheumatoid arthritis and in cancer. Here, we directly determined the binding site of the inverse agonist T140 on CXCR4 using photoaffinity labeling. Two T140 photoanalogs were synthesized containing the photoreactive amino acid p-benzoyl-l-phenylalanine (Bpa) in positions 5 or 10, yielding [Bpa(5)]T140 and [Bpa(10)]T140. Binding experiments on HEK293 cells stably expressing the wild-type CXCR4 receptor using 125I-SDF-1alpha demonstrated that T140 and both photoanalogs had affinities in the nanomolar range, similar to SDF-1alpha. Photolabeling led to the formation of specific, covalent 42 kDa T140-CXCR4 complexes. V8 protease digestion of both CXCR4/125I-[Bpa(5)]T140 and CXCR4/125I-[Bpa(10)]T140 adducts generated a fragment of 6kDa suggesting that the T140 photoanalogs labeled a fragment corresponding to Lys(154)-Glu(179) of the receptor's 4th transmembrane domain. Further digestion of this 6kDa fragment with endo Asp-N led to the generation of a shorter fragment validating the photolabeled region. Our results demonstrate that T140 interacts with residues of the fourth transmembrane domain of the CXCR4 receptor and provide new structural constraints enabling us to model the complex between T140 and CXCR4.


Assuntos
Benzofenonas/química , Oligopeptídeos/metabolismo , Fenilalanina/análogos & derivados , Marcadores de Fotoafinidade , Receptores CXCR4/metabolismo , Sequência de Aminoácidos , Fármacos Anti-HIV/química , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , DNA Viral/genética , HIV-1/genética , Humanos , Radioisótopos do Iodo , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Fenilalanina/química , Estrutura Terciária de Proteína , Provírus/genética , Receptores CXCR4/química , Relação Estrutura-Atividade
12.
J Immunol ; 183(3): 1759-66, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19592643

RESUMO

CCR2 is a chemokine receptor widely expressed by lymphomyeloid cells involved in maladaptive autoimmune ailments. Therefore CCR2 is of great interest as a biological target for immune suppression due to its direct implication in autoimmune diseases such as rheumatoid arthritis. We have generated a novel fusion protein using GM-CSF and an N-terminal truncated version of MCP-1/CCL2 (6-76, GMME1) and investigated its utility as a CCR2-specific immune suppressor. Using BRET studies, we found that distinct to CCL2, GMME1 binding to CCR2 led to altered conformational changes in the CCR2 homodimer and did not induce the recruitment of beta-arrestin 2 to the receptor. However, CCR2-dependent calcium mobilization, BAX induction and caspase-3 activation followed by cell death was observed. Using Th17 cells harvested from DBA/1 mice ill with bovine collagen-induced arthritis, we demonstrate that GMME1 is capable of blocking their production of IL-17 in vitro. Upon its delivery to mice symptomatic with inflammatory arthritis, a robust clinical recovery occurred with decreased paw thickness to normal levels and a significant reduction in anti-collagen Ab titer and rheumatoid factor titer, as well as reduction of proinflammatory cytokines levels both intraarticular and systemic. Our data demonstrate that GMME1 is a powerful synthetic suppressor cytokine that coopts CCR2-dependent cellular signaling and blunts the effects of CCR2-expressing lymphomyeloid cells causative of autoimmune arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Quimiocina CCL2/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Engenharia de Proteínas/métodos , Receptores CCR2/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Animais , Artrite Experimental/prevenção & controle , Células Cultivadas , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Interleucina-17 , Camundongos , Ligação Proteica , Receptores CCR2/química , Proteínas Recombinantes de Fusão/uso terapêutico , Subpopulações de Linfócitos T
13.
Cancer Res ; 69(14): 5793-800, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19584281

RESUMO

Induction of estrogen-regulated gene transcription by estrogen receptors ERalpha and ERbeta plays an important role in breast cancer development and growth. High expression of the chemokine receptor CXCR4 and its ligand CXCL12/stromal cell-derived factor 1 (SDF-1) has also been correlated with aggressive breast tumor phenotypes. Here, we describe a positive regulatory loop between the CXCR4/SDF-1 signaling pathway and ER transcriptional competence in human breast cancer cells. Treatment of breast carcinoma MCF-7 cells with SDF-1 increased ER transcriptional activity and expression of ER target genes, including SDF-1 itself. These effects were blocked by the antiestrogen ICI-182780 and by CXCR4 silencing and, conversely, estrogen-induced gene expression and growth of MCF-7 cells were impaired on CXCR4 inhibition. Both ERalpha and ERbeta were activated by SDF-1 in the presence of CXCR4 and by overexpression of a constitutively active CXCR4, indicating that CXCR4 signals to both receptors. In particular, ERbeta was able to translate the effects of SDF-1 on its own expression, as well as enhance activator protein 1 (AP-1) containing genes cyclin D1 and c-Myc in the presence of tamoxifen. This correlated with an increased ERbeta occupancy of responsive promoters at both estrogen-responsive and AP-1 elements. Ser-87, a conserved mitogen-activated protein kinase site in ERbeta, was highly phosphorylated by SDF-1, revealing an essential role of the AF-1 domain in response to CXCR4 activation. These results identify a complete autocrine loop between the CXCR4/SDF-1 and ERalpha/ERbeta signaling pathways that dictates ER-dependent gene expression and growth of breast cancer cells.


Assuntos
Quimiocina CXCL12/genética , Receptores CXCR4/genética , Receptores de Estrogênio/genética , Transdução de Sinais/genética , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina/genética , Serina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Fatores de Tempo , Fator de Transcrição AP-1/metabolismo
14.
Blood ; 113(21): 5176-85, 2009 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-19270265

RESUMO

Human T-cell lymphotropic virus type 1 (HTLV-1) entry involves the interaction between the surface (SU) subunit of the Env proteins and cellular receptor(s). Previously, our laboratories demonstrated that heparan sulfate proteoglycans (HSPGs) and neuropilin-1 (NRP-1), a receptor of VEGF(165), are essential for HTLV-1 entry. Here we investigated whether, as when binding VEGF(165), HSPGs and NRP-1 work in concert during HTLV-1 entry. VEGF(165) binds to the b domain of NRP-1 through both HSPG-dependent and -independent interactions, the latter involving its exon 8. We show that VEGF(165) is a selective competitor of HTLV-1 entry and that HTLV-1 mimics VEGF(165) to recruit HSPGs and NRP-1: (1) the NRP-1 b domain is required for HTLV-1 binding; (2) SU binding to target cells is blocked by the HSPG-binding domain of VEGF(165); (3) the formation of Env/NRP-1 complexes is enhanced by HSPGs; and (4) the HTLV SU contains a motif homologous to VEGF(165) exon 8. This motif directly binds to NRP-1 and is essential for HTLV-1 binding to, internalization into, and infection of CD4(+) T cells and dendritic cells. These findings demonstrate that HSPGs and NRP-1 function as HTLV-1 receptors in a cooperative manner and reveal an unexpected mimicry mechanism that may have major implications in vivo.


Assuntos
Proteoglicanas de Heparan Sulfato/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Neuropilina-1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ligação Viral , Ligação Competitiva , Células Cultivadas , Produtos do Gene env/metabolismo , Infecções por HTLV-I/virologia , Humanos , Mimetismo Molecular , Ligação Proteica , Receptores Virais/metabolismo
15.
J Immunol ; 180(10): 6977-87, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453620

RESUMO

IL-1 is a major proinflammatory cytokine which interacts with the IL-1 receptor I (IL-1RI) complex, composed of IL-1RI and IL-1R accessory protein subunits. Currently available strategies to counter pathological IL-1 signaling rely on a recombinant IL-1 receptor antagonist, which directly competes with IL-1 for its binding site. Presently, there are no small antagonists of the IL-1RI complex. Given this void, we derived 15 peptides from loops of IL-1R accessory protein, which are putative interactive sites with the IL-1RI subunit. In this study, we substantiate the merits of one of these peptides, rytvela (we termed "101.10"), as an inhibitor of IL-1R and describe its properties consistent with those of an allosteric negative modulator. 101.10 (IC(50) approximately 1 nM) blocked human thymocyte proliferation in vitro, and demonstrated robust in vivo effects in models of hyperthermia and inflammatory bowel disease as well as topically in contact dermatitis, superior to corticosteroids and IL-1ra; 101.10 did not bind to IL-1RI deficient cells and was ineffective in vivo in IL-1RI knockout mice. Importantly, characterization of 101.10, revealed noncompetitive antagonist actions and functional selectivity by blocking certain IL-1R pathways while not affecting others. Findings describe the discovery of a potent and specific small (peptide) antagonist of IL-1RI, with properties in line with an allosteric negative modulator.


Assuntos
Anti-Inflamatórios , Inflamação/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1 , Peptídeos/química , Peptídeos/farmacologia , Receptores de Interleucina-1/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Western Blotting , Dermatite/tratamento farmacológico , Humanos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Peptídeos/síntese química , Estrutura Terciária de Proteína , Ratos , Ratos Sprague-Dawley
16.
RNA ; 14(5): 981-91, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18367719

RESUMO

The human immunodeficiency virus of type 1 (HIV-1) uses a programmed -1 ribosomal frameshift to produce the precursor of its enzymes, and changes in frameshift efficiency reduce replicative fitness of the virus. We used a fluorescent two-reporter system to screen for peptides that reduce HIV-1 frameshift in bacteria, knowing that the frameshift can be reproduced in Escherichia coli. Expression of one reporter, the green fluorescent protein (GFP), requires the HIV-1 frameshift, whereas the second reporter, the red fluorescent protein (RFP), is used to assess normal translation. A peptide library biased for RNA binding was inserted into the sequence of the protein thioredoxin and expressed in reporter-containing bacteria, which were then screened by fluorescence-activated cell sorting (FACS). We identified peptide sequences that reduce frameshift efficiency by over 50% without altering normal translation. The identified sequences are also active against different frameshift stimulatory signals, suggesting that they bind a target important for frameshifting in general, probably the ribosome. Successful transfer of active sequences to a different scaffold in a eukaryotic test system demonstrates that the anti-frameshift activity of the peptides is neither due to scaffold-dependent conformation nor effects of the scaffold protein itself on frameshifting. The method we describe identifies peptides that will provide useful tools to further study the mechanism of frameshift and may permit the development of lead compounds of therapeutic interest.


Assuntos
Mudança da Fase de Leitura do Gene Ribossômico/efeitos dos fármacos , HIV-1/efeitos dos fármacos , HIV-1/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Sequência de Bases , Escherichia coli/genética , Escherichia coli/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , HIV-1/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Conformação de Ácido Nucleico , Biblioteca de Peptídeos , Peptídeos/genética , Plasmídeos/genética , Biossíntese de Proteínas/efeitos dos fármacos , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Vermelha Fluorescente
17.
Med Sci (Paris) ; 23(11): 980-4, 2007 Nov.
Artigo em Francês | MEDLINE | ID: mdl-18021711

RESUMO

CXCR4 is a clinically relevant chemokine receptor that has first gained attention as one of the cofactors for HIV entry into target cells. Moreover, the receptor is involved in cancer cell migration to distant metastatic sites and immune effector recruitment in inflammatory diseases such as asthma and rheumatoid arthritis. Unfortunately, pharmacologic intervention is complicated by the vital function of CXCR4 in the organism. The most prominent of these functions is its role in stem cell homing. The CXCR4 chemokine ligand, produced by bone marrow stromal cells, leads both to migration of hematopoietic stem cells towards this niche and their retention in this compartment. As models of G-protein coupled receptor (GPCR) activation evolve, it becomes clear that multiple factors modulate the functional outcome of ligand binding to a receptor. Modulation of GPCR activity, for example by allosteric ligands, may permit more subtle therapeutic approaches adapted to long term treatment. In addition, GPCR signalling can be altered by hetero-oligomerization of GPCRs. In this perspective, it might be possible to achieve modulation of GPCR signalling by also targeting the oligomerization partner of a given receptor. This approach is described using the example of strategies that aim at the optimization of stem cell homing in the context of cord blood-derived hematopoietic stem cell transplantation.


Assuntos
HIV/fisiologia , Receptores CXCR4/fisiologia , Movimento Celular , Humanos , Ligantes , Neoplasias/fisiopatologia , Ligação Proteica
18.
J Virol ; 80(14): 6844-54, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16809290

RESUMO

Human T-cell lymphotropic virus type 1 (HTLV-1) is transmitted through a viral synapse and enters target cells via interaction with the glucose transporter GLUT1. Here, we show that Neuropilin-1 (NRP1), the receptor for semaphorin-3A and VEGF-A165 and a member of the immune synapse, is also a physical and functional partner of HTLV-1 envelope (Env) proteins. HTLV-1 Env and NRP1 complexes are formed in cotransfected cells, and endogenous NRP1 contributes to the binding of HTLV-1 Env to target cells. NRP1 overexpression increases HTLV-1 Env-dependent syncytium formation. Moreover, overexpression of NRP1 increases both HTLV-1 and HTLV-2 Env-dependent infection, whereas down-regulation of endogenous NRP1 has the opposite effect. Finally, overexpressed GLUT1, NRP1, and Env form ternary complexes in transfected cells, and endogenous NRP1 and GLUT1 colocalize in membrane junctions formed between uninfected and HTLV-1-infected T cells. These data show that NRP1 is involved in HTLV-1 and HTLV-2 entry, suggesting that the HTLV receptor has a multicomponent nature.


Assuntos
Produtos do Gene env/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Infecções por HTLV-I/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Neuropilina-1/metabolismo , Receptores Virais/metabolismo , Animais , Células COS , Chlorocebus aethiops , Regulação para Baixo/genética , Produtos do Gene env/genética , Células Gigantes/metabolismo , Células Gigantes/virologia , Transportador de Glucose Tipo 1/genética , Infecções por HTLV-I/genética , Infecções por HTLV-I/transmissão , Infecções por HTLV-II/genética , Infecções por HTLV-II/metabolismo , Infecções por HTLV-II/transmissão , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 2 Humano/genética , Vírus Linfotrópico T Tipo 2 Humano/metabolismo , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neuropilina-1/genética , Fragmentos de Peptídeos/metabolismo , Receptores Virais/genética , Semaforina-3A/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
J Biol Chem ; 281(23): 16058-67, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16574649

RESUMO

Stimulation of freshly isolated rat hepatocytes with lysophosphatidic acid (LPA) resulted in LPA1 receptor-mediated and nitricoxide-dependent up-regulation of the immediate early genes iNOS (inducible nitric-oxide synthase (NOS)) and mPGES-1 (microsomal prostaglandin E synthase-1). Because LPA is a ligand for both cell surface and intracellular receptor sites and a potent endothelial NOS (eNOS) activator, we hypothesized that NO derived from activated nuclearized eNOS might participate in gene regulation. Herein we show, by confocal microscopy performed on porcine cerebral endothelial cells expressing native LPA1-receptor and eNOS and on HTC4 rat hepatoma cells co-transfected with recombinant human LPA1-receptor and fused eNOS-GFP cDNA, a dynamic eNOS translocation from peripheral to nuclear regions upon stimulation with LPA. Nuclear localization of eNOS and its downstream effector, soluble guanylate cyclase, were demonstrated in situ in rat liver specimens by immunogold labeling using specific antibodies. Stimulation of this nuclear fraction with LPA and the NO donor sodium nitroprusside resulted, respectively, in increased production of nitrite (and eNOS phosphorylation) and cGMP; these separate responses were also correspondingly blocked by NOS inhibitor L-NAME and soluble guanylate cyclase inhibitor ODQ. In addition, sodium nitroprusside evoked a sequential increase in nuclear Ca2+ transients, activation of p42 MAPK, NF-kappaB binding to DNA consensus sequence, and dependent iNOS RNA. This study describes a hitherto unrecognized molecular mechanism by which nuclear eNOS through ensuing NO modulates nuclear calcium homeostasis involved in gene transcription-associated events. Moreover, our findings strongly support the concept of the nucleus as an autonomous signaling compartment.


Assuntos
Genes Precoces , Inflamação/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Transdução de Sinais , Animais , Sequência de Bases , Núcleo Celular/metabolismo , Células Cultivadas , Primers do DNA , Expressão Gênica/efeitos dos fármacos , Guanilato Ciclase , Humanos , Fígado/metabolismo , Lisofosfolipídeos/farmacologia , Microscopia Confocal , Óxido Nítrico Sintase Tipo III/genética , Prostaglandina-E Sintases , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Guanilil Ciclase Solúvel , Suínos
20.
Arterioscler Thromb Vasc Biol ; 26(4): 744-50, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16439712

RESUMO

OBJECTIVE: Angiogenesis is essential physiologically in growth and pathologically in tumor development, chronic inflammatory disorders, and proliferative retinopathies. Activation of protease-activated receptor 2 (PAR2) leads to a proangiogenic response, but its mechanisms have yet to be specifically described. Here, we investigated the mode of action of PAR2 in retinal angiogenesis. METHODS AND RESULTS: PAR2-activating peptide, SLIGRL, increased retinal angiogenesis associated with an induction of vascular endothelial growth factor and angiopoetin-2 and most notably tie2 in the retina in vivo as well as in cultured neuroretinal endothelial cells. SLIGRL also induced release of the proinflammatory and angiogenic mediator tumor necrosis factor-alpha (TNF-alpha) via the MEK/extracellular signal-regulated kinase (ERK) (MEK/ERK) pathway in these endothelial cells. TNF-alpha, in turn, elicited tie2 expression by activating the MEK/ERK pathway. PAR2-evoked tie2 expression, endothelium proliferation (in vitro), and retinal neovascularization (in vivo) were abrogated by selective TNF-alpha blockers (neutralizing antibody infliximab and soluble TNF-alpha receptor-Fc fusion protein etanercept) as well as the MEK inhibitor PD98059. CONCLUSIONS: The proangiogenic properties of PAR2 are intertwined with its proinflammatory effects, such that in retinal vasculature, they depend on TNF-alpha and subsequent induction of tie2 via the MEK/ERK pathway.


Assuntos
Neovascularização Fisiológica , Receptor PAR-2/fisiologia , Angiopoietina-2/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Endotélio Vascular/fisiologia , Oligopeptídeos/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor TIE-2/metabolismo , Retina/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA