Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 107(6): 1123-1135, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32374043

RESUMO

Chemokines play critical roles in numerous physiologic and pathologic processes through their action on seven-transmembrane (TM) receptors. The N-terminal domain of chemokines, which is a key determinant of signaling via its binding within a pocket formed by receptors' TM helices, can be the target of proteolytic processing. An illustrative case of this regulatory mechanism is the natural processing of CXCL12 that generates chemokine variants lacking the first two N-terminal residues. Whereas such truncated variants behave as antagonists of CXCR4, the canonical G protein-coupled receptor of CXCL12, they are agonists of the atypical chemokine receptor 3 (ACKR3/CXCR7), suggesting the implication of different structural determinants in the complexes formed between CXCL12 and its two receptors. Recent analyses have suggested that the CXCL12 N-terminus first engages the TM helices of ACKR3 followed by the receptor N-terminus wrapping around the chemokine core. Here we investigated the first stage of ACKR3-CXCL12 interactions by comparing the activity of substituted or N-terminally truncated variants of CXCL12 toward CXCR4 and ACKR3. We showed that modification of the first two N-terminal residues of the chemokine (K1R or P2G) does not alter the ability of CXCL12 to activate ACKR3. Our results also identified the K1R variant as a G protein-biased agonist of CXCR4. Comparative molecular dynamics simulations of the complexes formed by ACKR3 either with CXCL12 or with the P2G variant identified interactions between the N-terminal 2-4 residues of CXCL12 and a pocket formed by receptor's TM helices 2, 6, and 7 as critical determinants for ACKR3 activation.


Assuntos
Quimiocina CXCL12/química , AMP Cíclico/química , Receptores CXCR4/química , Receptores CXCR/química , Sequência de Aminoácidos , Benzilaminas , Sítios de Ligação , Quimiocina CXCL11/química , Quimiocina CXCL11/genética , Quimiocina CXCL11/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ciclamos , AMP Cíclico/metabolismo , Expressão Gênica , Células HEK293 , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Humanos , Simulação de Dinâmica Molecular , Mutação , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
2.
Cell Chem Biol ; 25(4): 483-493.e9, 2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29478905

RESUMO

Neutrophils represent the most abundant immune cells recruited to inflamed tissues. A lack of dedicated tools has hampered their detection and study. We show that a synthesized peptide, MUB40, binds to lactoferrin, the most abundant protein stored in neutrophil-specific and tertiary granules. Lactoferrin is specifically produced by neutrophils among other leukocytes, making MUB40 a specific neutrophil marker. Naive mammalian neutrophils (human, guinea pig, mouse, rabbit) were labeled by fluorescent MUB40 conjugates (-Cy5, Dylight405). A peptidase-resistant retro-inverso MUB40 (RI-MUB40) was synthesized and its lactoferrin-binding property validated. Neutrophil lactoferrin secretion during in vitro Shigella infection was assessed with RI-MUB40-Cy5 using live cell microscopy. Systemically administered RI-MUB40-Cy5 accumulated at sites of inflammation in a mouse arthritis inflammation model in vivo and showed usefulness as a potential tool for inflammation detection using non-invasive imaging. Improving neutrophil detection with the universal and specific MUB40 marker will aid the study of broad ranges of inflammatory diseases.


Assuntos
Carbocianinas/química , Corantes Fluorescentes/química , Inflamação/diagnóstico , Lactoferrina/análise , Neutrófilos/imunologia , Peptídeos/química , Adulto , Animais , Biomarcadores/análise , Disenteria Bacilar/complicações , Disenteria Bacilar/diagnóstico , Disenteria Bacilar/imunologia , Disenteria Bacilar/microbiologia , Feminino , Cobaias , Humanos , Inflamação/complicações , Inflamação/imunologia , Inflamação/microbiologia , Lactoferrina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neutrófilos/microbiologia , Coelhos , Shigella/imunologia
3.
Open Biol ; 7(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29070611

RESUMO

Chemokines promote directional cell migration through binding to G-protein-coupled receptors, and as such are involved in a large array of developmental, homeostatic and pathological processes. They also interact with heparan sulfate (HS), the functional consequences of which depend on the respective location of the receptor- and the HS-binding sites, a detail that remains elusive for most chemokines. Here, to set up a biochemical framework to investigate how HS can regulate CXCL13 activity, we solved the solution structure of CXCL13. We showed that it comprises an unusually long and disordered C-terminal domain, appended to a classical chemokine-like structure. Using three independent experimental approaches, we found that it displays a unique association mode to HS, involving two clusters located in the α-helix and the C-terminal domain. Computational approaches were used to analyse the HS sequences preferentially recognized by the protein and gain atomic-level understanding of the CXCL13 dimerization induced upon HS binding. Starting with four sets of 254 HS tetrasaccharides, we identified 25 sequences that bind to CXCL13 monomer, among which a single one bound to CXCL13 dimer with high consistency. Importantly, we found that CXCL13 can be functionally presented to its receptor in a HS-bound form, suggesting that it can promote adhesion-dependent cell migration. Consistently, we designed CXCL13 mutations that preclude interaction with HS without affecting CXCR5-dependent cell signalling, opening the possibility to unambiguously demonstrate the role of HS in the biological function of this chemokine.


Assuntos
Sítios de Ligação , Quimiocina CXCL13/química , Quimiocina CXCL13/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/metabolismo , Conformação Molecular , Domínios e Motivos de Interação entre Proteínas , Sequência de Aminoácidos , Quimiocina CXCL13/genética , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Multimerização Proteica , Estabilidade Proteica , Proteínas Recombinantes , Soluções , Relação Estrutura-Atividade
4.
J Biol Chem ; 292(19): 7784-7794, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28325837

RESUMO

Old long-lived proteins contain dehydroalanine (Dha) and dehydrobutyrine (Dhb), two amino acids engendered by dehydration of serines and threonines, respectively. Although these residues have a suspected role in protein cross-linking and aggregation, their direct implication has yet to be determined. Here, we have taken advantage of the ability of the enteropathogen Shigella to convert the phosphothreonine residue of the pT-X-pY consensus sequence of ERK and p38 into Dhb and followed the impact of dehydration on the fate of the two MAPKs. To that end, we have generated the first antibodies recognizing Dhb-modified proteins and allowing tracing them as they form. We showed that Dhb modifications accumulate in a long-lasting manner in Shigella-infected cells, causing subsequent formation of covalent cross-links of MAPKs. Moreover, the Dhb signal correlates precisely with the activation of the Shigella type III secretion apparatus, thus evidencing injectisome activity. This observation is the first to document a causal link between Dhb formation and protein cross-linking in live cells. Detection of eliminylation is a new avenue to phosphoproteome regulation in eukaryotes that will be instrumental for the development of type III secretion inhibitors.


Assuntos
Proteínas de Bactérias/metabolismo , Carbono-Oxigênio Liases/metabolismo , Sistema de Sinalização das MAP Quinases , Shigella/enzimologia , Treonina/química , Alanina/análogos & derivados , Alanina/química , Aminobutiratos/química , Animais , Anticorpos/química , Células CACO-2 , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HeLa , Humanos , Camundongos , Ligação Proteica , Proteômica , Especificidade por Substrato , Sistemas de Secreção Tipo III , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Virol ; 91(8)2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28122982

RESUMO

Anti-human immunodeficiency virus type 1 (HIV-1) nonneutralizing antibodies (nnAbs) capable of antibody-dependent cellular cytotoxicity (ADCC) have been identified as a protective immune correlate in the RV144 vaccine efficacy trial. Broadly neutralizing antibodies (bNAbs) also mediate ADCC in cell culture and rely on their Fc region for optimal efficacy in animal models. Here, we selected 9 monoclonal nnAbs and 5 potent bNAbs targeting various epitopes and conformations of the gp120/41 complex and analyzed the potency of the two types of antibodies to bind and eliminate HIV-1-infected cells in culture. Regardless of their neutralizing activity, most of the selected antibodies recognized and killed cells infected with two laboratory-adapted HIV-1 strains. Some nnAbs also bound bystander cells that may have captured viral proteins. However, in contrast to the bNAbs, the nnAbs bound poorly to reactivated infected cells from 8 HIV-positive individuals and did not mediate effective ADCC against these cells. The nnAbs also inefficiently recognize cells infected with 8 different transmitted-founder (T/F) isolates. The addition of a synthetic CD4 mimetic enhanced the binding and killing efficacy of some of the nnAbs in an epitope-dependent manner without reaching the levels achieved by the most potent bNAbs. Overall, our data reveal important qualitative and quantitative differences between nnAbs and bNAbs in their ADCC capacity and strongly suggest that the breadth of recognition of HIV-1 by nnAbs is narrow.IMPORTANCE Most of the anti-HIV antibodies generated by infected individuals do not display potent neutralizing activities. These nonneutralizing antibodies (nnAbs) with antibody-dependent cellular cytotoxicity (ADCC) have been identified as a protective immune correlate in the RV144 vaccine efficacy trial. However, in primate models, the nnAbs do not protect against simian-human immunodeficiency virus (SHIV) acquisition. Thus, the role of nnAbs with ADCC activity in protection from infection remains debatable. In contrast, broadly neutralizing antibodies (bNAbs) neutralize a large array of viral strains and mediate ADCC in cell culture. We analyzed the capacities of 9 nnAbs and 5 bNAbs to eliminate infected cells. We selected 18 HIV-1 strains, including virus reactivated from the reservoir of HIV-positive individuals and transmitted-founder isolates. We report that the nnAbs bind poorly to cells infected with primary HIV-1 strains and do not mediate potent ADCC. Overall, our data show that the breadth of recognition of HIV-1 by nnAbs is narrow.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/imunologia , Anticorpos Monoclonais/imunologia , Células Cultivadas , Humanos
6.
Sci Signal ; 9(452): ra107, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27803285

RESUMO

Chemokines stimulate signals in cells by binding to G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors. These chemoattractant cytokines also interact with heparan sulfate (HS), which provides positional information within tissues in the form of haptotactic gradients along which cells can migrate directionally. To investigate the mechanism by which HS modulates chemokine functions, we used the CXC chemokine CXCL12, which exists in different isoforms that all signal through CXCR4 but have distinct HS-binding domains. In experiments with both cell-associated and solubilized CXCR4, we found that although CXCL12γ bound to CXCR4 with a higher affinity than did CXCL12α, CXCL12γ displayed reduced signaling and chemotactic activities. These properties were caused by the specific carboxyl-terminal region of CXCL12γ, which, by interacting with CXCR4 sulfotyrosines, mediated high-affinity, but nonproductive, binding to CXCR4. HS prevented CXCL12γ from interacting with the CXCR4 sulfotyrosines, thereby functionally presenting the chemokine to its receptor such that its activity was similar to that of CXCL12α. HS had no effects on the binding of CXCL12α to CXCR4 or its biological activity, suggesting that this polysaccharide controls CXCL12 in an isoform-specific manner. These data suggest that the HS-dependent regulation of chemokine functions extends beyond the simple process of immobilization and directly modulates receptor ligation and activation.


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Heparitina Sulfato/metabolismo , Receptores CXCR4/metabolismo , Linfócitos T/metabolismo , Humanos , Isoformas de Proteínas/metabolismo , Linfócitos T/citologia
7.
Sci Rep ; 6: 34829, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27721488

RESUMO

The CD4 and the cryptic coreceptor binding sites of the HIV-1 envelope glycoprotein are key to viral attachment and entry. We developed new molecules comprising a CD4 mimetic peptide linked to anionic compounds (mCD4.1-HS12 and mCD4.1-PS1), that block the CD4-gp120 interaction and simultaneously induce the exposure of the cryptic coreceptor binding site, rendering it accessible to HS12- or PS1- mediated inhibition. Using a cynomolgus macaque model of vaginal challenge with SHIV162P3, we report that mCD4.1-PS1, formulated into a hydroxyethyl-cellulose gel provides 83% protection (5/6 animals). We next engineered the mCD4 moiety of the compound, giving rise to mCD4.2 and mCD4.3 that, when conjugated to PS1, inhibited cell-free and cell-associated HIV-1 with particularly low IC50, in the nM to pM range, including some viral strains that were resistant to the parent molecule mCD4.1. These chemically defined molecules, which target major sites of vulnerability of gp120, are stable for at least 48 hours in conditions replicating the vaginal milieu (37 °C, pH 4.5). They efficiently mimic several large gp120 ligands, including CD4, coreceptor or neutralizing antibodies, to which their efficacy compares very favorably, despite a molecular mass reduced to 5500 Da. Together, these results support the development of such molecules as potential microbicides.


Assuntos
Fármacos Anti-HIV/farmacologia , Antígenos CD4/química , HIV-1/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Administração Intravaginal , Animais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/química , Linfócitos T CD4-Positivos/virologia , Estabilidade de Medicamentos , Feminino , Géis/administração & dosagem , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , HIV-1/patogenicidade , Heparitina Sulfato/química , Humanos , Macaca fascicularis , Mimetismo Molecular , Peptídeos/farmacocinética , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/patogenicidade , Vagina/virologia
8.
Biomaterials ; 35(32): 8903-15, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25088726

RESUMO

Glycosaminoglycans (GAGs) are ubiquitously present at the cell surface and in extracellular matrix, and crucial for matrix assembly, cell-cell and cell-matrix interactions. The supramolecular presentation of GAG chains, along with other matrix components, is likely to be functionally important but remains challenging to control and to characterize, both in vivo and in vitro. We present a method to create well-defined biomimetic surfaces that display GAGs, either alone or together with other cell ligands, in a background that suppresses non-specific binding. Through the design of the immobilization platform - a streptavidin monolayer serves as a molecular breadboard for the attachment of various biotinylated ligands - and a set of surface-sensitive in situ analysis techniques (including quartz crystal microbalance and spectroscopic ellipsometry), the biomimetic surfaces are tailor made with tight control on biomolecular orientation, surface density and lateral mobility. Analysing the interactions between a selected GAG (heparan sulphate, HS) and the HS-binding chemokine CXCL12α (also called SDF-1α), we demonstrate that these surfaces are versatile for biomolecular and cellular interaction studies. T-lymphocytes are found to adhere specifically to surfaces presenting CXCL12α, both when reversibly bound through HS and when irreversibly immobilized on the inert surface, even in the absence of any bona fide cell adhesion ligand. Moreover, surfaces which present both HS-bound CXCL12α and the intercellular adhesion molecule 1 (ICAM-1) synergistically promote cell adhesion. Our surface biofunctionalization strategy should be broadly applicable for functional studies that require a well-defined supramolecular presentation of GAGs along with other matrix or cell-surface components.


Assuntos
Biomimética/métodos , Membrana Celular/química , Quimiocina CXCL12/química , Glicosaminoglicanos/química , Molécula 1 de Adesão Intercelular/química , Biotinilação , Adesão Celular , Matriz Extracelular/química , Fibronectinas/química , Heparitina Sulfato/química , Humanos , Células Jurkat , Ligantes , Modelos Moleculares , Ligação Proteica , Proteínas Recombinantes/química , Soroalbumina Bovina/química , Estreptavidina/química , Ressonância de Plasmônio de Superfície , Propriedades de Superfície , Linfócitos T/química
9.
Int J Oncol ; 43(3): 775-84, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23817837

RESUMO

There is substantial evidence suggesting that certain parasites can have antitumor properties. We evaluated mucin peptides derived from the helminth Echinococcus granulosus (denominated Egmuc) as potential inducers of antitumor activity. We present data showing that Egmuc peptides were capable of inducing an increase of activated NK cells in the spleen of immunized mice, a fact that was correlated with the capacity of splenocytes to mediate killing of tumor cells. We demonstrated that Egmuc peptides enhance LPS-induced maturation of dendritic cells in vitro by increasing the production of IL-12p40p70 and IL-6 and that Egmuc-treated DCs may activate NK cells, as judged by an increased expression of CD69. This evidence may contribute to the design of tumor vaccines and open new horizons in the use of parasite-derived molecules in the fight against cancer.


Assuntos
Antineoplásicos/administração & dosagem , Mucinas/administração & dosagem , Neoplasias/tratamento farmacológico , Peptídeos/administração & dosagem , Animais , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Echinococcus granulosus/química , Humanos , Subunidade p40 da Interleucina-12/imunologia , Interleucina-6/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Lectinas Tipo C/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Mucinas/química , Mucinas/isolamento & purificação , Neoplasias/imunologia , Neoplasias/patologia , Peptídeos/química , Peptídeos/isolamento & purificação , Baço/efeitos dos fármacos , Baço/imunologia
10.
PLoS One ; 7(6): e37873, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22675496

RESUMO

Glaucoma, the most common cause of irreversible blindness, is a neuropathy commonly initiated by pathological ocular hypertension due to unknown mechanisms of trabecular meshwork degeneration. Current antiglaucoma therapy does not target the causal trabecular pathology, which may explain why treatment failure is often observed. Here we show that the chemokine CXCL12, its truncated form SDF-1(5-67), and the receptors CXCR4 and CXCR3 are expressed in human glaucomatous trabecular tissue and a human trabecular cell line. SDF-1(5-67) is produced under the control of matrix metallo-proteinases, TNF-α, and TGF-ß2, factors known to be involved in glaucoma. CXCL12 protects in vitro trabecular cells from apoptotic death via CXCR4 whereas SDF-1(5-67) induces apoptosis through CXCR3 and caspase activation. Ocular administration of SDF-1(5-67) in the rat increases intraocular pressure. In contrast, administration of a selective CXCR3 antagonist in a rat model of ocular hypertension decreases intraocular pressure, prevents retinal neurodegeneration, and preserves visual function. The protective effect of CXCR3 antagonism is related to restoration of the trabecular function. These data demonstrate that proteolytic cleavage of CXCL12 is involved in trabecular pathophysiology, and that local administration of a selective CXCR3 antagonist may be a beneficial therapeutic strategy for treating ocular hypertension and subsequent retinal degeneration.


Assuntos
Quimiocina CXCL12/farmacologia , Hipertensão Ocular/complicações , Hipertensão Ocular/fisiopatologia , Receptores CXCR3/antagonistas & inibidores , Degeneração Retiniana/complicações , Degeneração Retiniana/prevenção & controle , Malha Trabecular/fisiopatologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular , Citoproteção/efeitos dos fármacos , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Glaucoma/complicações , Glaucoma/metabolismo , Glaucoma/patologia , Glaucoma/fisiopatologia , Humanos , Pressão Intraocular/efeitos dos fármacos , Masculino , Ratos , Ratos Long-Evans , Receptores CXCR3/metabolismo , Receptores CXCR4/metabolismo , Degeneração Retiniana/fisiopatologia , Estresse Fisiológico/efeitos dos fármacos , Malha Trabecular/efeitos dos fármacos , Malha Trabecular/patologia , Visão Ocular/efeitos dos fármacos
11.
J Biol Chem ; 287(19): 15916-22, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22427651

RESUMO

Imaging living cells and organs requires innovative, specific, efficient, and well tolerated fluorescent markers targeting cellular components. Such tools will allow proceeding to the dynamic analysis of cells and the adaptation of tissues to environmental cues. In this study, we have identified and synthesized a novel non-toxic fluorescent marker allowing a specific fluorescent staining of the human colonic mucus. Our strategy to identify a molecule able to specifically bind to the human colonic mucus was on the basis of the mucus adhesion properties of commensal bacteria. We identified and characterized the mucus-binding property of a 70-amino acid domain (MUB(70)) expressed on the surface of Lactobacillus strains. The chemical synthesis of MUB(70) was achieved using the human commensal bacterium Lactobacillus reuteri AF120104 protein as a template. The synthesized Cy5-conjugated MUB(70) marker specifically stained the colonic mucus on fixed human, rabbit, and guinea pig tissues. Interestingly, murine tissue was not stained, suggesting significant differences in the composition of the murine colonic mucus. In addition, this marker stained the mucus of living cultured human colonic cells (HT29-MTX) and human colonic tissue explants. Using a biotinylated derivative of MUB(70), we demonstrated that this peptide binds specifically to Muc2, the most abundant secreted mucin, through its glycosylated moieties. Hence, Cy5-MUB(70) is a novel and specific fluorescent marker for mammalian colonic mucus. It may be used for live imaging analysis but also, as demonstrated in this study, as a marker for the diagnosis and the prognosis of colonic mucinous carcinomas.


Assuntos
Proteínas de Bactérias/metabolismo , Colo/metabolismo , Limosilactobacillus reuteri/metabolismo , Mucina-2/metabolismo , Muco/metabolismo , Sequência de Aminoácidos , Animais , Aderência Bacteriana , Proteínas de Bactérias/química , Proteínas de Bactérias/farmacologia , Sobrevivência Celular , Colo/microbiologia , Eletroforese em Gel de Poliacrilamida , Corantes Fluorescentes/química , Glicosilação , Cobaias , Células HT29 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/fisiologia , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Muco/microbiologia , Ligação Proteica , Coelhos , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
12.
Chem Biol ; 19(1): 131-9, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22284360

RESUMO

The HIV-1 envelope gp120, which features both the virus receptor (CD4) and coreceptor (CCR5/CXCR4) binding sites, offers multiple sites for therapeutic intervention. However, the latter becomes exposed, thus vulnerable to inhibition, only transiently when the virus has already bound cellular CD4. To pierce this defense mechanism, we engineered a series of heparan sulfate mimicking tridecapeptides and showed that one of them target the gp120 coreceptor binding site with µM affinity. Covalently linked to a CD4-mimetic that binds to gp120 and renders the coreceptor binding domain available to be targeted, the conjugated tridecapeptide now displays nanomolar affinity for its target. Using solubilized coreceptors captured on top of sensorchip we show that it inhibits gp120 binding to both CCR5 and CXCR4 and in peripheral blood mononuclear cells broadly inhibits HIV-1 replication with an IC(50) of 1 nM.


Assuntos
Fármacos Anti-HIV/química , Antígenos CD4/metabolismo , HIV-1/efeitos dos fármacos , Heparitina Sulfato/química , Peptídeos/química , Sequência de Aminoácidos , Fármacos Anti-HIV/síntese química , Fármacos Anti-HIV/farmacologia , Sítios de Ligação , Antígenos CD4/química , Membrana Celular/metabolismo , Proteína gp120 do Envelope de HIV/química , Proteína gp120 do Envelope de HIV/metabolismo , Humanos , Peptídeos/síntese química , Ligação Proteica , Receptores CCR5/química , Receptores CCR5/metabolismo , Receptores CXCR4/química , Receptores CXCR4/metabolismo , Replicação Viral/efeitos dos fármacos
13.
Biochem Pharmacol ; 82(9): 1163-74, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21803029

RESUMO

Aberrant and constitutive NF-κB activation are frequently reported in numerous tumor types, making its inhibition an attractive target for the treatment of certain cancers. NEMO (NF-κB essential modulator) is the crucial component of the canonical NF-κB pathway that mediates IκB kinase (IKK) complex activation. IKK activation resides in the ability of the C-terminal domain of NEMO to properly dimerize and interact with linear and K63-linked polyubiquitin chains. Here, we have identified a new NEMO peptide inhibitor, termed UBI (ubiquitin binding inhibitor) that derives from the NOA/NUB/UBAN ubiquitin binding site located in the CC2-LZ domain of NEMO. UBI specifically inhibits the NF-κB pathway at the IKK level in different cell types stimulated by a variety of NF-κB signals. Circular dichroïsm and fluorescence studies showed that UBI exhibits an increased α-helix character and direct, good-affinity binding to the NOA-LZ region of NEMO. We also showed that UBI targets NEMO in cells but its mode of inhibition is completely different from the previously reported LZ peptide (herein denoted NOA-LZ). UBI does not promote dissociation of NEMO subunits in cells but impairs the interaction between the NOA UBD of NEMO and polyubiquitin chains. Importantly, we showed that UBI efficiently competes with the in vitro binding of K63-linked chains, but not with linear chains. The identification of this new NEMO inhibitor emphasizes the important contribution of K63-linked chains for IKK activation in NF-κB signaling and would provide a new tool for studying the complex role of NF-κB in inflammation and cancer.


Assuntos
Quinase I-kappa B/metabolismo , NF-kappa B/metabolismo , Ubiquitina/metabolismo , Sítios de Ligação , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/genética , Modelos Moleculares , NF-kappa B/antagonistas & inibidores , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
14.
Leukemia ; 25(8): 1286-1296, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21494253

RESUMO

Steady-state egress of hematopoietic progenitor cells can be rapidly amplified by mobilizing agents such as AMD3100, the mechanism, however, is poorly understood. We report that AMD3100 increased the homeostatic release of the chemokine stromal cell derived factor-1 (SDF-1) to the circulation in mice and non-human primates. Neutralizing antibodies against CXCR4 or SDF-1 inhibited both steady state and AMD3100-induced SDF-1 release and reduced egress of murine progenitor cells over mature leukocytes. Intra-bone injection of biotinylated SDF-1 also enhanced release of this chemokine and murine progenitor cell mobilization. AMD3100 directly induced SDF-1 release from CXCR4(+) human bone marrow osteoblasts and endothelial cells and activated uPA in a CXCR4/JNK-dependent manner. Additionally, ROS inhibition reduced AMD3100-induced SDF-1 release, activation of circulating uPA and mobilization of progenitor cells. Norepinephrine treatment, mimicking acute stress, rapidly increased SDF-1 release and progenitor cell mobilization, whereas ß2-adrenergic antagonist inhibited both steady state and AMD3100-induced SDF-1 release and progenitor cell mobilization in mice. In conclusion, this study reveals that SDF-1 release from bone marrow stromal cells to the circulation emerges as a pivotal mechanism essential for steady-state egress and rapid mobilization of hematopoietic progenitor cells, but not mature leukocytes.


Assuntos
Células da Medula Óssea/metabolismo , Quimiocina CXCL12/fisiologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Norepinefrina/farmacologia , Receptores CXCR4/fisiologia , Animais , Benzilaminas , Células Cultivadas , Ciclamos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células Estromais/metabolismo
15.
J Pept Sci ; 16(2): 98-104, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20014324

RESUMO

This work contributes to highlight the benefits of pseudoproline dipeptides introduction in difficult SPPS. We show how a slight modification in the positioning choice conditioned the synthesis achievement of a 54 amino acid long caveolin-1 peptide encompassing the intramembrane domain. Furthermore, we report a side reaction correlated with the coupling steps and generating truncated fragments with a mass deviation of + 42 Da. Considering the need of structural data for membrane proteins, most of which are considered as prevalent therapeutic targets, chemical synthesis provides an interesting alternative pathway to obtain hydrophobic domains by pushing back the frontiers of conventional RP methods of purification.


Assuntos
Caveolina 1/síntese química , Dipeptídeos , Proteínas de Membrana/química , Prolina/análogos & derivados , Tiazóis/química , Sequência de Aminoácidos , Caveolina 1/química , Cromatografia Líquida de Alta Pressão , Dipeptídeos/química , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Prolina/química
16.
Nat Chem Biol ; 5(10): 743-8, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19734912

RESUMO

The HIV-1 envelope, gp120, which features the binding determinants for both CD4 and coreceptor recognition, is key for virus entry and represents an attractive pharmacological target. However, critical domains for entry (coreceptor and CD4 binding sites) are either cryptic or located in partially occluded cavities. Here we developed a chemical approach to synthesize a CD4-mimetic peptide linked to a heparan sulfate dodecasaccharide. This molecule binds to gp120, induces the exposure of the coreceptor binding domain and renders it available for interaction with the oligosaccharide. The linkage between the CD4 mimetic and the heparan sulfate derivative provides strong cooperative effects, resulting in low-nanomolar antiviral activity toward both CCR5- and CXCR4-tropic HIV-1 strains. This compound, which has the unique ability to simultaneously target two critical and highly conserved regions of gp120, establishes a new type of inhibitor and suggests a general concept for the inhibition of numerous other biological systems.


Assuntos
Fármacos Anti-HIV/farmacologia , Antígenos CD4/farmacologia , Glicoconjugados/farmacologia , HIV-1/efeitos dos fármacos , Heparitina Sulfato/farmacologia , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Fármacos Anti-HIV/química , Sítios de Ligação , Antígenos CD4/química , Glicoconjugados/química , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/fisiologia , Heparitina Sulfato/química , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/virologia , Modelos Moleculares
17.
Glycobiology ; 19(12): 1511-24, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19717493

RESUMO

We have recently reported that the CXC-chemokine stromal cell-derived factor-1 (SDF-1)/CXCL12 induces proliferation, migration, and invasion of the Huh7 human hepatoma cells through its G-protein-coupled receptor CXCR4 and that glycosaminoglycans (GAGs) are involved in these events. Here, we demonstrate by surface plasmon resonance that the chemokine binds to GAG mimetics obtained by grafting carboxylate, sulfate or acetate groups onto a dextran backbone. We also demonstrate that chemically modified dextrans inhibit SDF-1/CXCL12-mediated in vitro chemotaxis and anchorage-independent cell growth in a dose-dependent manner. The binding of GAG mimetics to the chemokine and their effects in modulating the SDF-1/CXCL12 biological activities are mainly related to the presence of sulfate groups. Furthermore, the mRNA expression of enzymes involved in heparan sulfate biosynthesis, such as exostosin-1 and -2 or N-deacetylase N-sulfotransferases remained unchanged, but heparanase mRNA and protein expressions in Huh7 cells were decreased upon GAG mimetic treatment. Moreover, decreasing heparanase-1 mRNA levels by RNA interference significantly reduced SDF-1/CXCL12-induced extracellular signal-regulated kinase 1/2 (ERK 1/2) phosphorylation. Therefore, we suggest that GAG mimetic effects on SDF-1/CXCL12-mediated hepatoma cell chemotaxis may rely on decreased heparanase expression, which impairs SDF-1/CXCL12's signaling. Altogether, these data suggest that GAG mimetics may compete with cellular heparan sulfate chains for the binding to SDF-1/CXCL12 and may affect heparanase expression, leading to reduced SDF-1/CXCL12 mediated in vitro chemotaxis and growth of hepatoma cells.


Assuntos
Carcinoma Hepatocelular/patologia , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/antagonistas & inibidores , Glicosaminoglicanos/farmacologia , Neoplasias Hepáticas/patologia , Ligação Competitiva/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Adesão Celular/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacologia , Glucuronidase/antagonistas & inibidores , Glucuronidase/biossíntese , Glicosaminoglicanos/agonistas , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Invasividade Neoplásica , RNA Interferente Pequeno/farmacologia , Células Tumorais Cultivadas
18.
Biochim Biophys Acta ; 1790(12): 1643-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19695308

RESUMO

BACKGROUND: In addition to their physiologic effects in inflammation and angiogenesis, chemokines are involved in cancer pathology. The CXC-chemokine stromal cell-derived factor-1 (SDF-1)/CXCL12 mediates its biological activities through activation of G protein-coupled receptor CXCR4 and binds to glycosaminoglycans (GAGs). METHODS: Using Bio-coat cell migration chambers, specific antagonists, flow cytometry and RNA interference, we evaluate the involvement of heparan sulfate proteoglycans (HSPG) in the SDF-1/CXCL12-induced invasion of human cervix epitheloid carcinoma HeLa cells. RESULTS: The SDF-1/CXCL12-induced cell invasion is dependent on CXCR4. Furthermore, Protein Kinase C delta (PKC delta) and c-jun NH2-terminal kinase/stress-activated protein kinase (JNK/SAPK) are implicated in this event, but not extracellular signal-regulated kinase (ERK) 1/2. Moreover, the invasion of HeLa cells induced by SDF-1/CXCL12 was dependent on matrix metalloproteinase-9 (MMP-9). The pre-incubation of HeLa cells with heparin or with anti-heparan sulfate antibodies or with beta-d-xyloside inhibited SDF-1/CXCL12-mediated cell invasion. Furthermore, the down-regulation of syndecan-4, a heparan sulfate proteoglycan, decreased SDF-1/CXCL12-mediated HeLa cell invasion. GAGs, probably on syndecan-4, are involved in SDF-1/CXCL12-mediated cell chemotaxis. GENERAL SIGNIFICANCE: These data suggest that targeting the glycosaminoglycan/chemokine interaction could be a new therapeutic approach for carcinomas in which SDF-1/CXCL12 is involved.


Assuntos
Carcinoma in Situ/patologia , Quimiocina CXCL12/farmacologia , Glicosaminoglicanos/fisiologia , Sindecana-4/fisiologia , Neoplasias do Colo do Útero/patologia , Antracenos/farmacologia , Benzilaminas , Carcinoma in Situ/metabolismo , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/fisiologia , Ciclamos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Células HeLa , Compostos Heterocíclicos/farmacologia , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/fisiologia , Invasividade Neoplásica , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo
19.
Blood ; 113(24): 6085-93, 2009 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-19380869

RESUMO

The stromal cell-derived factor-1/CXCL12 chemokine engages the CXCR4 and CXCR7 receptors and regulates homeostatic and pathologic processes, including organogenesis, leukocyte homeostasis, and tumorigenesis. Both receptors are widely expressed in mammalian cells, but how they cooperate to respond to CXCL12 is not well understood. Here, we show that CXCR7 per se does not trigger G(alphai) protein-dependent signaling, although energy transfer assays indicate that it constitutively interacts with G(alphai) proteins and undergoes CXCL12-mediated conformational changes. Moreover, when CXCR4 and CXCR7 are coexpressed, we show that receptor heterodimers form as efficiently as receptor homodimers, thus opening the possibility that CXCR4/CXCR7 heterodimer formation has consequences on CXCL12-mediated signals. Indeed, expression of CXCR7 induces conformational rearrangements within preassembled CXCR4/G(alphai) protein complexes and impairs CXCR4-promoted G(alphai)-protein activation and calcium responses. Varying CXCR7 expression levels and blocking CXCL12/CXCR7 interactions in primary T cells suggest that CXCR4/CXCR7 heterodimers form in primary lymphocytes and regulate CXCL12-promoted chemotaxis. Taken together, these results identify CXCR4/CXCR7 heterodimers as distinct functional units with novel properties, which can contribute to the functional plasticity of CXCL12.


Assuntos
Quimiocina CXCL12/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Multimerização Proteica , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Citometria de Fluxo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Rim/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo
20.
Glycobiology ; 18(12): 1054-64, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18796646

RESUMO

Chemokine stromal cell-derived factor-1 (SDF-1) is a potent chemoattractant involved in leukocyte trafficking and metastasis. Heparan sulfate on the cell surface binds SDF-1 and may modulate its function as a coreceptor of this chemokine. A major effect of the glycosaminoglycan binding may be on the quaternary structure of SDF-1, which has been controversially reported as a monomer or a dimer. We have investigated the effect of sulfated oligosaccharides on the oligomerization of SDF-1 and of its mutated form SDF-1 (3/6), using affinity capillary electrophoresis (ACE) hyphenated to mass spectrometry (MS). Coupled to MS, ACE allowed the study for the first time of the effect of size-defined oligosaccharides on the quaternary organization of SDF-1 in muM range concentrations, i.e., lower values than the mM values previously reported in NMR, light scattering, and ultracentrifugation experiments. Our results showed that in the absence of sulfated oligosaccharides, SDF-1 is mostly monomeric in solution. However, dimer formation was observed upon interaction with heparin-sulfated oligosaccharides despite the mM Kd values for dimerization. A SDF-1/oligosaccharide 2/1 complex was detected, indicating that oligosaccharide binding promoted the dimerization of SDF-1. Heparin tetrasaccharide but not disaccharide promoted dimer formation, suggesting that the dimer required to be stabilized by a long enough bound oligosaccharide. The SDF-1/oligosaccharide 1/1 complex was only observed with heparin disaccharide and fucoidan pentasaccharide, pointing out the role of specific structural determinants in promoting dimer formation. These results underline the importance of dimerization induced by glycosaminoglycans for chemokine functionality.


Assuntos
Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Heparina/química , Polissacarídeos/química , Sítios de Ligação , Configuração de Carboidratos , Linhagem Celular Tumoral , Movimento Celular , Dimerização , Eletroforese Capilar , Heparina/metabolismo , Humanos , Espectrometria de Massas , Modelos Moleculares , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , Conformação Proteica , Espectrometria de Massas por Ionização por Electrospray
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...