Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(3)2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799570

RESUMO

Background: The atypical chemokine receptor 3 (ACKR3) belongs to the superfamily of G protein-coupled receptors (GPCRs). Unlike classical GPCRs, this receptor does not activate G proteins in most cell types but recruits ß-arrestins upon activation. ACKR3 plays an important role in cancer and vascular diseases. As recruitment of ß-arrestins is triggered by phosphorylation of the C-terminal tail of GPCRs, we studied the role of different potential phosphorylation sites within the ACKR3 C-tail to further delineate the molecular mechanism of internalization and trafficking of this GPCR. Methods: We used various bioluminescence and fluorescence resonance energy transfer-based sensors and techniques in Human Embryonic Kidney (HEK) 293T cells expressing WT or phosphorylation site mutants of ACKR3 to measure CXCL12-induced recruitment of ß-arrestins and G-protein-coupled receptor kinases (GRKs), receptor internalization and trafficking. Results: Upon CXCL12 stimulation, ACKR3 recruits both ß-arrestin 1 and 2 with equivalent kinetic profiles. We identified interactions with GRK2, 3 and 5, with GRK2 and 3 being important for ß-arrestin recruitment. Upon activation, ACKR3 internalizes and recycles back to the cell membrane. We demonstrate that ß-arrestin recruitment to the receptor is mainly determined by a single cluster of phosphorylated residues on the C-tail of ACKR3, and that residue T352 and in part S355 are important residues for ß-arrestin1 recruitment. Phosphorylation of the C-tail appears essential for ligand-induced internalization and important for differential ß-arrestin recruitment. GRK2 and 3 play a key role in receptor internalization. Moreover, ACKR3 can still internalize when ß-arrestin recruitment is impaired or in the absence of ß-arrestins, using alternative internalization pathways. Our data indicate that distinct residues within the C-tail of ACKR3 differentially regulate CXCL12-induced ß-arrestin recruitment, ACKR3 trafficking and internalization.


Assuntos
Endocitose , Receptores CXCR/metabolismo , beta-Arrestina 1/metabolismo , beta-Arrestina 2/metabolismo , Técnicas Biossensoriais , Quimiocina CXCL12/farmacologia , Transferência Ressonante de Energia de Fluorescência , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Quinase 3 de Receptor Acoplado a Proteína G/metabolismo , Células HEK293 , Humanos , Cinética , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Receptores CXCR/agonistas , Receptores CXCR/genética , beta-Arrestina 1/genética , beta-Arrestina 2/genética
2.
Sci Rep ; 11(1): 3426, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33564089

RESUMO

Most seven transmembrane receptors (7TMRs) are G protein-coupled receptors; however, some 7TMRs evoke intracellular signals through ß-arrestin as a biased receptor. As several ß-arrestin-biased agonists have been reported to be cardioprotective, we examined the role of the chemokine receptor CXCR7 as a ß-arrestin-biased receptor in the heart. Among 510 7TMR genes examined, Cxcr7 was the most abundantly expressed in the murine heart. Single-cell RNA-sequencing analysis revealed that Cxcr7 was abundantly expressed in cardiomyocytes and fibroblasts. Cardiomyocyte-specific Cxcr7 null mice showed more prominent cardiac dilatation and dysfunction than control mice 4 weeks after myocardial infarction. In contrast, there was no difference in cardiac phenotypes between fibroblast-specific Cxcr7-knockout mice and control mice even after myocardial infarction. TC14012, a specific agonist of CXCR7, significantly recruited ß-arrestin to CXCR7 in CXCR7-expressing cells and activated extracellular signal-regulated kinase (ERK) in neonatal rat cardiomyocytes. Cxcr7 expression was significantly increased and ERK was activated in the border zone of the heart in control, but not Cxcr7 null mice. These results indicate that the abundantly expressed CXCR7 in cardiomyocytes may play a protective role in the heart as a ß-arrestin-biased receptor and that CXCR7 may be a novel therapeutic target for myocardial infarction.


Assuntos
Sistema de Sinalização das MAP Quinases , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptores CXCR/metabolismo , beta-Arrestina 1/metabolismo , Animais , Camundongos , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Oligopeptídeos/farmacologia , Receptores CXCR/agonistas , Receptores CXCR/genética , beta-Arrestina 1/genética
3.
Nat Commun ; 11(1): 4855, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32978390

RESUMO

The atypical chemokine receptor 3 (ACKR3) plays a pivotal role in directing the migration of various cellular populations and its over-expression in tumors promotes cell proliferation and invasiveness. The intracellular signaling pathways transducing ACKR3-dependent effects remain poorly characterized, an issue we addressed by identifying the interactome of ACKR3. Here, we report that recombinant ACKR3 expressed in HEK293T cells recruits the gap junction protein Connexin 43 (Cx43). Cx43 and ACKR3 are co-expressed in mouse brain astrocytes and human glioblastoma cells and form a complex in embryonic mouse brain. Functional in vitro studies show enhanced ACKR3 interaction with Cx43 upon ACKR3 agonist stimulation. Furthermore, ACKR3 activation promotes ß-arrestin2- and dynamin-dependent Cx43 internalization to inhibit gap junctional intercellular communication in primary astrocytes. These results demonstrate a functional link between ACKR3 and gap junctions that might be of pathophysiological relevance.


Assuntos
Astrócitos/metabolismo , Comunicação Celular/fisiologia , Conexina 43/metabolismo , Junções Comunicantes/patologia , Receptores CXCR/metabolismo , Animais , Proliferação de Células , Conexina 43/efeitos dos fármacos , Conexinas/metabolismo , Técnicas de Introdução de Genes , Glioblastoma/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Domínios e Motivos de Interação entre Proteínas , Receptores CXCR/agonistas , Receptores CXCR/genética , Transdução de Sinais/fisiologia
4.
Mol Pharmacol ; 96(6): 737-752, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548340

RESUMO

The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.


Assuntos
Receptores CXCR4/fisiologia , Receptores CXCR/fisiologia , Sequência de Aminoácidos , Animais , Benzilaminas , Ciclamos , Compostos Heterocíclicos/metabolismo , Compostos Heterocíclicos/farmacologia , Humanos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores CXCR/agonistas , Receptores CXCR/antagonistas & inibidores , Receptores CXCR4/agonistas , Receptores CXCR4/antagonistas & inibidores
5.
Brain Behav Immun ; 82: 319-337, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31505255

RESUMO

High mobility group box 1 (HMGB1) is an endogenous alarmin that drives the pathogenesis of neurodegenerative disorders including cognitive decline. Therefore, HMGB1 is thought to be a common biomarker as well as promising therapeutic target for neuroinflammation associated with neurocognitive disorders. Here, for the first time, we have unmasked the potential inhibitory effect of a novel receptor of HMGB1-CXCL12 complex; atypical chemokine receptor 3 (ACKR3/CXCR7) on HMGB1 induced glial phenotype switching, neuroinflammation, and subsequent memory loss. Upregulation of CXCR7 inhibits HMGB1-CXCL12 complex induced peripheral immune cells infiltration to CNS by regulating blood-brain barrier (BBB) integrity in HMGB1 induced dementia model of mice. Whereas, gene knockdown study by RNA interference (non-invasive intranasal delivery to animal model) shows CXCR7 ablation aggravates inflammatory responses in hippocampus region and immune cell infiltration to CNS tissue by breached BBB. This study also indicates the important role of CXCR7 molecule in maintaining CNS homeostasis by balancing M1/M2 microglia, A1/A2 astrocytes, long term potentiation/long term depression markers which ultimately ameliorates HMGB1 induced neurodegeneration, synaptic depression and memory loss (assessed by both radial arm maze and Morris water maze) in male mice model of dementia. Overall, the study summarizes several significant protective functions afforded by CXCR7 against HMGB1 induced disbalance in neuroimmunological axis, neurodegeneration and memory loss and thereby provides a new paradigm for strategic development of novel therapeutics against neurodegenerative diseases with dementia as a common symptom.


Assuntos
Alarminas/farmacologia , Transtornos da Memória/metabolismo , Receptores CXCR/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Quimiocina CXCL12/metabolismo , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Feminino , Masculino , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/patologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neuroglia/metabolismo , Neuroimunomodulação/fisiologia , Neuroproteção , Receptores CXCR/agonistas , Receptores CXCR4/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais
6.
PLoS One ; 13(9): e0204041, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30248140

RESUMO

We observed in PRESTO-Tango ß-arrestin recruitment assays that the α1-adrenergic receptor (AR) antagonist prazosin activates chemokine (C-X-C motif) receptor (CXCR)4. This prompted us to further examine this unexpected pharmacological behavior. We screened a panel of 14 α1/2- and ß1/2/3-AR antagonists for CXCR4 and atypical chemokine receptor (ACKR)3 agonist activity in PRESTO-Tango assays against the cognate agonist CXCL12. We observed that multiple α1-AR antagonists activate CXCR4 (CXCL12 = prazosin = cyclazosin > doxazosin) and ACKR3 (CXCL12 = prazosin = cyclazosin > alfuzosin = doxazosin = phentolamine > terazosin = silodosin = tamsulosin). The two strongest CXCR4/ACKR3 activators, prazosin and cyclazosin, were selected for a more detailed evaluation. We found that the drugs dose-dependently activate both receptors in ß-arrestin recruitment assays, stimulate ERK1/2 phosphorylation in HEK293 cells overexpressing each receptor, and that their effects on CXCR4 could be inhibited with AMD3100. Both α1-AR antagonists induced significant chemical shift changes in the 1H-13C-heteronuclear single quantum correlation spectrum of CXCR4 and ACKR3 in membranes, suggesting receptor binding. Furthermore, prazosin and cyclazosin induced internalization of endogenous CXCR4/ACKR3 in human vascular smooth muscle cells (hVSMC). While these drugs did not in induce chemotaxis in hVSMC, they inhibited CXCL12-induced chemotaxis with high efficacy and potency (IC50: prazosin-4.5 nM, cyclazosin 11.6 pM). Our findings reveal unexpected pharmacological properties of prazosin, cyclazosin, and likely other α1-AR antagonists. The results of the present study imply that prazosin and cyclazosin are biased or partial CXCR4/ACKR3 agonists, which function as potent CXCL12 antagonists. Our findings could provide a mechanistic basis for previously observed anti-cancer properties of α1-AR antagonists and support the concept that prazosin could be re-purposed for the treatment of disease processes in which CXCR4 and ACKR3 are thought to play significant pathophysiological roles, such as cancer metastases or various autoimmune pathologies.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Receptores CXCR4/agonistas , Receptores CXCR/agonistas , Sítios de Ligação , Células Cultivadas , Quimiocina CXCL12/antagonistas & inibidores , Quimiotaxia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Ressonância Magnética Nuclear Biomolecular , Prazosina/farmacologia , Quinazolinas/farmacologia , Quinoxalinas/farmacologia , Receptores CXCR/química , Receptores CXCR4/química , beta-Arrestinas/metabolismo
7.
Biochem Pharmacol ; 153: 299-309, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29530506

RESUMO

The atypical chemokine receptor ACKR3/CXCR7 plays crucial roles in numerous physiological processes but also in viral infection and cancer. ACKR3 shows strong propensity for activation and, unlike classical chemokine receptors, can respond to chemokines from both the CXC and CC families as well as to the endogenous peptides BAM22 and adrenomedullin. Moreover, despite belonging to the G protein coupled receptor family, its function appears to be mainly dependent on ß-arrestin. ACKR3 has also been shown to continuously cycle between the plasma membrane and the endosomal compartments, suggesting a possible role as a scavenging receptor. So far, the molecular basis accounting for these atypical binding and signalling properties remains elusive. Noteworthy, ACKR3 extracellular domains bear three disulphide bridges. Two of them lie on top of the two main binding subpockets and are conserved among chemokine receptors, and one, specific to ACKR3, forms an intra-N terminus four-residue-loop of so far unknown function. Here, by mutational and functional studies, we examined the impact of the different disulphide bridges for ACKR3 folding, ligand binding and activation. We showed that, in contrast to most classical chemokine receptors, none of the extracellular disulphide bridges was essential for ACKR3 function. However, the disruption of the unique ACKR3 N-terminal loop drastically reduced the binding of CC chemokines whereas it only had a mild impact on CXC chemokine binding. Mutagenesis also uncovered that chemokine and endogenous non-chemokine ligands interact and activate ACKR3 according to distinct binding modes characterized by different transmembrane domain subpocket occupancy and N-terminal loop contribution, with BAM22 mimicking the binding mode of CC chemokine N terminus.


Assuntos
Quimiocinas CXC/metabolismo , Análise Mutacional de DNA/métodos , Líquido Extracelular/metabolismo , Mutação/fisiologia , Receptores CXCR/agonistas , Receptores CXCR/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Quimiocinas CXC/genética , Humanos , Ligação Proteica , Receptores CXCR/genética
8.
J Med Chem ; 60(23): 9653-9663, 2017 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-29045152

RESUMO

The chemokine receptor CXCR7 is an attractive target for a variety of diseases. While several small-molecule modulators of CXCR7 have been reported, peptidic macrocycles may provide advantages in terms of potency, selectivity, and reduced off-target activity. We produced a series of peptidic macrocycles that incorporate an N-linked peptoid functionality where the peptoid group enabled us to explore side-chain diversity well beyond that of natural amino acids. At the same time, theoretical calculations and experimental assays were used to track and reduce the polarity while closely monitoring the physicochemical properties. This strategy led to the discovery of macrocyclic peptide-peptoid hybrids with high CXCR7 binding affinities (Ki < 100 nM) and measurable passive permeability (Papp > 5 × 10-6 cm/s). Moreover, bioactive peptide 25 (Ki = 9 nM) achieved oral bioavailability of 18% in rats, which was commensurate with the observed plasma clearance values upon intravenous administration.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Peptoides/química , Peptoides/farmacologia , Receptores CXCR/agonistas , Receptores CXCR/metabolismo , Administração Oral , Animais , Disponibilidade Biológica , Cães , Humanos , Compostos Macrocíclicos/administração & dosagem , Compostos Macrocíclicos/química , Compostos Macrocíclicos/farmacocinética , Compostos Macrocíclicos/farmacologia , Células Madin Darby de Rim Canino , Masculino , Simulação de Acoplamento Molecular , Peptídeos/administração & dosagem , Peptídeos/farmacocinética , Peptoides/administração & dosagem , Peptoides/farmacocinética , Ratos , Ratos Wistar
9.
Arch Toxicol ; 91(1): 325-338, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27136898

RESUMO

2,3,7,8 Tetrachlorodibenzo-p-dioxin (TCDD) is an aromatic, long-lived environmental contaminant. While the pathogenesis of TCDD-induced toxicity is poorly understood, it has been shown that the aryl hydrocarbon receptor (AHR) is required. However, the specific transcriptomic changes that lead to toxic outcomes have not yet been identified. We previously identified a panel of 33 genes that respond to TCDD treatment in two TCDD-sensitive rodent species. To identify genes involved in the onset of hepatic toxicity, we explored 25 of these in-depth using liver from two rat strains: the TCDD-resistant Han/Wistar (H/W) and the TCDD-sensitive Long-Evans (L-E). Time course and dose-response analyses of mRNA abundance following TCDD insult indicate that eight genes are similarly regulated in livers of both strains of rat, suggesting that they are not central to the severe L-E-specific TCDD-induced toxicities. The remaining 17 genes exhibited various divergent mRNA abundances between L-E and H/W strains after TCDD treatment. Several genes displayed a biphasic response where the initial response to TCDD treatment was followed by a secondary response, usually of larger magnitude in L-E liver. This secondary response was most often an exaggeration of the original TCDD-induced response. Only cytochrome b5 type A (microsomal) (Cyb5a) had equivalent TCDD sensitivity to the prototypic AHR-responsive cytochrome P450, family 1, subfamily a, polypeptide 1 (Cyp1a1), while six genes were less sensitive. Four genes showed an early inter-strain difference that was sustained throughout most of the time course (atypical chemokine receptor 3 (Ackr3), collagen, type XVIII, alpha 1 (Col18a1), Cyb5a and glutamate dehydrogenase 1 (Glud1)), and of those genes examined in this study, are most likely to represent genes involved in the pathogenesis of TCDD-induced hepatotoxicity in L-E rats.


Assuntos
Carcinógenos Ambientais/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , RNA Mensageiro/metabolismo , Animais , Animais não Endogâmicos , Carcinógenos Ambientais/administração & dosagem , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Colágeno Tipo VIII/agonistas , Colágeno Tipo VIII/antagonistas & inibidores , Colágeno Tipo VIII/genética , Colágeno Tipo VIII/metabolismo , Citocromos b5/antagonistas & inibidores , Citocromos b5/química , Citocromos b5/genética , Citocromos b5/metabolismo , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Feminino , Perfilação da Expressão Gênica , Glutamato Desidrogenase , Cinética , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Dibenzodioxinas Policloradas/administração & dosagem , Ratos Long-Evans , Receptores CXCR/agonistas , Receptores CXCR/antagonistas & inibidores , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores de Glutamato/química , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo
10.
Biochem Pharmacol ; 114: 14-21, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27238288

RESUMO

The atypical chemokine receptor CXCR7/ACKR3 binds two endogenous chemokines, CXCL12 and CXCL11, and is upregulated in many cancers or following infection by several cancer-inducing viruses, including HHV-8. ACKR3 is a ligand-scavenging receptor and does not activate the canonical G protein pathways but was proposed to trigger ß-arrestin-dependent signaling. Here, we identified the human herpesvirus 8-encoded CC chemokine vCCL2/vMIP-II as a third high-affinity ligand for ACKR3. vCCL2 acted as partial ACKR3 agonist, inducing ß-arrestin recruitment to the receptor, subsequent reduction of its surface levels and its delivery to endosomes. In addition, ACKR3 reduced vCCL2-triggered MAP kinase and PI3K/Akt signaling through other chemokine receptors. Our data suggest that ACKR3 acts as a scavenger receptor for vCCL2, regulating its availability and activity toward human receptors, thereby likely controlling its function in HHV-8 infection. Our study provides new insights into the complex crosstalk between viral chemokines and host receptors as well as into the biology of ACKR3, this atypical and still enigmatic receptor.


Assuntos
Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Herpesvirus Humano 8/metabolismo , Receptores CXCR/agonistas , Animais , Ligação Competitiva , Células CHO , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Quimiocina CXCL12/metabolismo , Quimiocinas/genética , Cricetulus , Endossomos/metabolismo , Citometria de Fluxo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Humanos , Ligantes , Receptor Cross-Talk , Receptores CXCR/genética , Transfecção , beta-Arrestina 2/metabolismo
11.
Nat Med ; 22(2): 154-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26779814

RESUMO

Although the lung can undergo self-repair after injury, fibrosis in chronically injured or diseased lungs can occur at the expense of regeneration. Here we study how a hematopoietic-vascular niche regulates alveolar repair and lung fibrosis. Using intratracheal injection of bleomycin or hydrochloric acid in mice, we show that repetitive lung injury activates pulmonary capillary endothelial cells (PCECs) and perivascular macrophages, impeding alveolar repair and promoting fibrosis. Whereas the chemokine receptor CXCR7, expressed on PCECs, acts to prevent epithelial damage and ameliorate fibrosis after a single round of treatment with bleomycin or hydrochloric acid, repeated injury leads to suppression of CXCR7 expression and recruitment of vascular endothelial growth factor receptor 1 (VEGFR1)-expressing perivascular macrophages. This recruitment stimulates Wnt/ß-catenin-dependent persistent upregulation of the Notch ligand Jagged1 (encoded by Jag1) in PCECs, which in turn stimulates exuberant Notch signaling in perivascular fibroblasts and enhances fibrosis. Administration of a CXCR7 agonist or PCEC-targeted Jag1 shRNA after lung injury promotes alveolar repair and reduces fibrosis. Thus, targeting of a maladapted hematopoietic-vascular niche, in which macrophages, PCECs and perivascular fibroblasts interact, may help to develop therapy to spur lung regeneration and alleviate fibrosis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Capilares/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lesão Pulmonar/metabolismo , Pulmão/metabolismo , Proteínas de Membrana/metabolismo , Fibrose Pulmonar/metabolismo , Receptores CXCR/metabolismo , Regeneração/fisiologia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Capilares/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Fibroblastos/efeitos dos fármacos , Fibrose , Imunofluorescência , Humanos , Ácido Clorídrico/toxicidade , Proteína Jagged-1 , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Oligopeptídeos/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Circulação Pulmonar/efeitos dos fármacos , Circulação Pulmonar/fisiologia , RNA Interferente Pequeno/farmacologia , Receptores CXCR/agonistas , Receptores Notch/metabolismo , Regeneração/efeitos dos fármacos , Proteínas Serrate-Jagged , Proteína Smad3/efeitos dos fármacos , Proteína Smad3/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Via de Sinalização Wnt
12.
Curr Top Med Chem ; 16(13): 1441-51, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26369824

RESUMO

Under physiological conditions, CXCL12 modulates cell proliferation, survival, angiogenesis, and migration mainly through CXCR4. Interestingly, the newly discovered receptor CXCR7 for CXCL12 is highly expressed in many tumor cells as well as tumor-associated blood vessels, although the level of CXCR7 in normal blood cells is low. Recently, many studies have suggested that CXCR7 promotes cell growth and metastasis in various cancers, including lymphoma and leukemia, hepatocecullar, ovarian, colorectal, breast and lung cancer. Compared to CXCR4, CXCR7 is a non-classical GPCR that is unable to activate G proteins. The function of CXCR7 is generally considered to be mediated by: (a) recruiting ß-arrestin-2; (b) heterodimerizing with CXCR4; and (c) acting as a "scavenger" of CXCL12, thus lowering the level of CXCL12 to weaken the activity of CXCR4. However, the crosstalk between CXCL12/CXCR7/CXCR4 and other signaling pathways (such as the p38 MAPK pathway, the PI3K/mTOR pathway, the STAT3 signaling, and metalloproteinases MMP-9 and MMP-2) is more complicated. The function of CXCR7 is also involved in modulating tumor microenvironment, tumor cell migration and apoptosis. Understanding these complex interactions will provide insight in drug design targeting the CXCR7 as potential anticancer therapy.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CXCL12/metabolismo , Desenho de Fármacos , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/química , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quimiocina CXCL12/antagonistas & inibidores , Humanos , Receptores CXCR/agonistas , Receptores CXCR/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores
13.
Biochemistry ; 54(7): 1505-15, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25669416

RESUMO

The chemokine receptor CXCR7 is an atypical CXCL12 receptor that, as opposed to the classical CXCL12 receptor CXCR4, signals preferentially via the ß-arrestin pathway and does not mediate chemotaxis. We previously reported that the cyclic peptide TC14012, a potent CXCR4 antagonist, also engaged CXCR7, albeit with lower potency. Surprisingly, the compound activated the CXCR7-arrestin pathway. The reason underlying the opposite effects of TC14012 on CXCR4 and CXCR7, and the mode of binding of TC14012 to CXCR7, remained unclear. The mode of binding of TC14012 to CXCR4 is known from cocrystallization of its analogue CVX15 with CXCR4. We here report the the mode of binding of TC14012 to CXCR7 by combining the use of compound analogues, receptor mutants, and molecular modeling. We find that the mode of binding of TC14012 to CXCR7 is indeed similar to that of CVX15 to CXCR4, with compound positions Arg2 and Arg14 engaging CXCR7 key residues D179(4.60) (on the tip of transmembrane domain 4) and D275(6.58) (on the tip of transmembrane domain 6), respectively. Interestingly, the TC14012 parent compound T140 is not a CXCR7 agonist, because of conformational constraints in its pharmacophore, which in TC14012 are relieved through C-terminal amidation. However, an engineered salt bridge between the CXCR7 ECL2 substitution R197D and compound residue Arg1 permitted T140 agonism by repositioning the compound in the binding pocket. In conclusion, our results show that the opposite effect of TC14012 on CXCR4 and CXCR7 is not explained by different binding modes. Rather, engagement of the interface between transmembrane domains and extracellular loops readily triggers CXCR7, but not CXCR4, activation.


Assuntos
Oligopeptídeos/farmacologia , Receptores CXCR/agonistas , Receptores CXCR/metabolismo , Sequência de Aminoácidos , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Oligopeptídeos/química , Receptores CXCR/química , Receptores CXCR4/antagonistas & inibidores
14.
Mol Med ; 20: 435-47, 2014 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-25032954

RESUMO

Chemokine (C-X-C motif) receptor (CXCR) 4 and atypical chemokine receptor (ACKR) 3 ligands have been reported to modulate cardiovascular function in various disease models. The underlying mechanisms, however, remain unknown. Thus, it was the aim of the present study to determine how pharmacological modulation of CXCR4 and ACKR3 regulate cardiovascular function. In vivo administration of TC14012, a CXCR4 antagonist and ACKR3 agonist, caused cardiovascular collapse in normal animals. During the cardiovascular stress response to hemorrhagic shock, ubiquitin, a CXCR4 agonist, stabilized blood pressure, whereas coactivation of CXCR4 and ACKR3 with CXC chemokine ligand 12 (CXCL12), or blockade of CXCR4 with AMD3100 showed opposite effects. While CXCR4 and ACKR3 ligands did not affect myocardial function, they selectively altered vascular reactivity upon α1-adrenergic receptor (AR) activation in pressure myography experiments. CXCR4 activation with ubiquitin enhanced α1-AR-mediated vasoconstriction, whereas ACKR3 activation with various natural and synthetic ligands antagonized α1-AR-mediated vasoconstriction. The opposing effects of CXCR4 and ACKR3 activation by CXCL12 could be dissected pharmacologically. CXCR4 and ACKR3 ligands did not affect vasoconstriction upon activation of voltage-operated Ca(2+) channels or endothelin receptors. Effects of CXCR4 and ACKR3 agonists on vascular α1-AR responsiveness were independent of the endothelium. These findings suggest that CXCR4 and ACKR3 modulate α1-AR reactivity in vascular smooth muscle and regulate hemodynamics in normal and pathological conditions. Our observations point toward CXCR4 and ACKR3 as new pharmacological targets to control vasoreactivity and blood pressure.


Assuntos
Receptores Adrenérgicos alfa 1/fisiologia , Receptores CXCR4/fisiologia , Receptores CXCR/fisiologia , Agonistas Adrenérgicos/farmacologia , Animais , Benzilaminas , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Quimiocina CXCL12/farmacologia , Ciclamos , Compostos Heterocíclicos/farmacologia , Técnicas In Vitro , Ligantes , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Oligopeptídeos/farmacologia , Fenilefrina/farmacologia , Ratos Endogâmicos Lew , Receptores CXCR/agonistas , Receptores CXCR4/agonistas , Receptores CXCR4/antagonistas & inibidores , Choque Hemorrágico/fisiopatologia , Ubiquitina/farmacologia , Vasoconstrição/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos
15.
Ophthalmic Res ; 52(1): 17-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24853648

RESUMO

AIMS: To observe the effect of TC14012 (a CXCR4 antagonist and CXCR7 agonist) on alkali burn-induced corneal neovascularization (CNV) in a mouse model. METHODS: CNV was induced in vivo by alkali burns on the corneas of BALB/c mice. A total of 54 mice treated with alkali burns were randomly divided into 3 groups, each of which received one of the following treatments: bilateral subconjunctival injections of TC14012 for 3 consecutive days, bilateral subconjunctival injections of balanced saline (BS) for 3 consecutive days or no treatment (blank control). The areas of CNV were measured on days 3, 7 and 14 after the alkali burns. CXCR4, CXCR7, vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMP) mRNAs were detected and quantified by real-time reverse transcription PCR on days 7 and 14. Additionally, the expression of the proteins CXCR4, CXCR7, VEGF, ß-arrestin 2, total ERK1/2 and phospho-ERK1/2 was determined by Western blotting. RESULTS: On day 7 after the alkali burns, the CNV area, VEGF, MMP-2 and MMP-9 mRNA levels, and VEGF, ß-arrestin 2 and phospho-ERK1/2 protein levels were increased in the TC14012 group compared with the nontreatment and BS groups. However, on day 14, the CNV area, CXCR4, CXCR7, VEGF, MMP-2 and MMP-9 mRNA levels, and the CXCR4, CXCR7, VEGF and ß-arrestin 2 protein levels were significantly decreased in the TC14012 group. CONCLUSIONS: TC14012 initially enhanced alkali burn-induced CNV but reduced CNV in later stages. In addition to CXCR4, CXCR7 is involved in the pathogenesis of CNV.


Assuntos
Queimaduras Químicas/tratamento farmacológico , Neovascularização da Córnea/tratamento farmacológico , Modelos Animais de Doenças , Queimaduras Oculares/induzido quimicamente , Oligopeptídeos/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR/agonistas , Animais , Queimaduras Químicas/etiologia , Queimaduras Químicas/patologia , Túnica Conjuntiva/efeitos dos fármacos , Neovascularização da Córnea/induzido quimicamente , Neovascularização da Córnea/patologia , Regulação da Expressão Gênica/fisiologia , Injeções Intraoculares , Masculino , Metaloproteinases da Matriz/genética , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores CXCR/genética , Receptores CXCR4/genética , Hidróxido de Sódio , Fator A de Crescimento do Endotélio Vascular/genética
16.
Circulation ; 129(11): 1244-53, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24374972

RESUMO

BACKGROUND: The aim of this study was to determine the role of the chemokine receptor CXCR7 in atherosclerosis and vascular remodeling. CXCR7 is the alternative receptor of CXCL12, which regulates stem cell-mediated vascular repair and limits atherosclerosis via its receptor, CXCR4. METHODS AND RESULTS: Wire-induced injury of the carotid artery was performed in mice with a ubiquitous, conditional deletion of CXCR7 and in mice treated with the synthetic CXCR7 ligand CCX771. The effect of CCX771 treatment on atherosclerosis was studied in apolipoprotein E-deficient (Apoe(-/-)) mice fed a high-fat diet for 12 weeks. Lipoprotein fractions were quantified in the plasma of Apoe(-/-) mice by fast protein liquid chromatography. Uptake of DiI-labeled very low-density lipoprotein to adipose tissue was determined by 2-photon microscopy. We show that genetic deficiency of Cxcr7 increased neointima formation and lesional macrophage accumulation in hyperlipidemic mice after vascular injury. This was related to increased serum cholesterol levels and subsequent hyperlipidemia-induced monocytosis. Conversely, administration of the CXCR7 ligand CCX771 to Apoe(-/-) mice inhibited lesion formation and ameliorated hyperlipidemia after vascular injury and during atherosclerosis. Treatment with CCX771 reduced circulating very low-density lipoprotein levels but not low-density lipoprotein or high-density lipoprotein levels and increased uptake of very low-density lipoprotein into Cxcr7-expressing white adipose tissue. This effect of CCX771 was associated with an enhanced lipase activity and reduced expression of Angptl4 in adipose tissue. CONCLUSIONS: CXCR7 regulates blood cholesterol by promoting its uptake in adipose tissue. This unexpected cholesterol-lowering effect of CXCR7 is beneficial for atherosclerotic vascular diseases, presumably via amelioration of hyperlipidemia-induced monocytosis, and can be augmented with a synthetic CXCR7 ligand.


Assuntos
Tecido Adiposo/metabolismo , Aterosclerose/metabolismo , Colesterol/metabolismo , Hiperlipidemias/metabolismo , Receptores CXCR/biossíntese , Animais , Aterosclerose/prevenção & controle , Hiperlipidemias/prevenção & controle , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR/agonistas
17.
Biochem Biophys Res Commun ; 431(4): 772-6, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23333329

RESUMO

The CXCL12/CXCR4 axis is involved in many cellular responses for host homeostasis, and malfunction of this signaling pathway is associated with a variety of diseases. It is now known that CXCL12 also binds to another newly identified chemokine receptor, CXCR7, which does not couple with a G-protein. CXCR7 can form homodimers, or heterodimers with CXCR4, and is believed to sequester the chemokine CXCL12, although the CXCL12/CXCR7 axis activates MAP kinases through ß-arrestin. Therefore, it has not been well defined how CXCR7 activation affects CXCL12-induced cellular events. To elucidate the function of CXCR7, we prepared CXCR7 agonist Compound 1. Compound 1 is a selective and potent CXCR7 agonist that clearly has the activity to recruit ß-arrestin toward CXCR7. It also activates MAP kinases Akt and ERK. Using this compound, we confirmed that the CXCR7 agonist, but not an antagonistic antibody, did inhibit CXCL12 induced HUVEC tube formation, suggesting that activation of CXCR7 ameliorates CXCL12 induced cellular events, probably by affecting on CXCR4 function. We show that ß-arrestin recruitment to CXCR4 is reduced by over-expression of CXCR7 and activation of CXCR7 by agonist treatment reduces the protein level of CXCR4. Based on our results, together with reported information, we propose that CXCR7, when up-regulated upon inflammation, can act as a negative regulator of CXCR4 by heterodimerizing with CXCR4, inducing its internalization and degradation. This mechanism suggests that CXCR7 agonists can have a therapeutic effect on CXCL12 causing diseases by countering the effects of CXCL12.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Piridinas/farmacologia , Quinolonas/farmacologia , Receptores CXCR4/metabolismo , Receptores CXCR/agonistas , Regulação para Baixo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Piridinas/química , Quinolonas/química
18.
Eur J Med Chem ; 51: 184-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22424612

RESUMO

The chemokine receptor CXCR7 is an atypical G protein-coupled receptor as it preferentially signals through the ß-arrestin pathway rather than through G proteins. CXCR7 is thought to be of importance in cancer and the development of CXCR7-targeting ligands is of huge importance to further elucidate the pharmacology and the therapeutic potential of CXCR7. In the present study, we synthesized 24 derivatives based on a compound scaffold patented by Chemocentryx and obtained CXCR7 ligands with pK(i) values ranging from 5.3 to 8.1. SAR studies were supported by computational 3D Fingerprint studies, revealing several important affinity descriptors. Two key compounds (29 and 30, VUF11207 and VUF11403) were found to be high-potency ligands that induce recruitment of ß-arrestin2 and subsequent internalization of CXCR7, making them important tool compounds in future CXCR7 research.


Assuntos
Amidas/química , Técnicas de Química Sintética , Modelos Moleculares , Receptores CXCR/agonistas , Estireno/química , Estireno/farmacologia , Células HEK293 , Humanos , Conformação Molecular , Relação Quantitativa Estrutura-Atividade , Estireno/síntese química
19.
Mol Pharmacol ; 75(5): 1240-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19255243

RESUMO

The bicyclam AMD3100 is known as a small synthetic inhibitor of the CXCL12-binding chemokine receptor CXCR4. Here, we show that AMD3100 also binds to the alternative CXCL12 receptor CXCR7. CXCL12 or AMD3100 alone activate beta-arrestin recruitment to CXCR7, which we identify as a previously unreported signaling pathway of CXCR7. In addition, AMD3100 increases CXCL12 binding to CXCR7 and CXCL12-induced conformational rearrangements in the receptor dimer as measured by bioluminescence resonance energy transfer. Moreover, small but reproducible increases in the potency of CXCL12-induced arrestin recruitment to CXCR7 by AMD3100 are observed. Taken together, our data suggest that AMD3100 is an allosteric agonist of CXCR7. The finding that AMD3100 not only binds CXCR4, but also to CXCR7, with opposite effects on the two receptors, calls for caution in the use of the compound as a tool to dissect CXCL12 effects on the respective receptors in vitro and in vivo.


Assuntos
Compostos Heterocíclicos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR/agonistas , Regulação Alostérica , Arrestinas/metabolismo , Benzilaminas , Células Cultivadas , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacologia , Ciclamos , Dimerização , Humanos , Luminescência , Receptores CXCR/química , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...