Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 403
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(30): e2404000121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39008676

RÉSUMÉ

Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon ß-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of ß-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.


Sujet(s)
Simulation de dynamique moléculaire , Liaison aux protéines , Récepteurs CXCR , Humains , Récepteurs CXCR/métabolisme , Récepteurs CXCR/génétique , Sites de fixation , Conformation des protéines , bêta-Arrestine 1/métabolisme , bêta-Arrestine 1/génétique , Ligands , Cellules HEK293 , Mutagenèse dirigée , Régulation allostérique , Relation structure-activité
2.
Cells ; 13(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38920657

RÉSUMÉ

The reciprocal modulation between the CXCL12/CXCR4/ACKR3 axis and the STAT3 signaling pathway plays a crucial role in the progression of various diseases and neoplasms. Activation of the CXCL12/CXCR4/ACKR3 axis triggers the STAT3 pathway through multiple mechanisms, while the STAT3 pathway also regulates the expression of CXCL12. This review offers a thorough and systematic analysis of the reciprocal regulatory mechanisms between the CXCL12/CXCR4/ACKR3 signaling axis and the STAT3 signaling pathway in the context of diseases, particularly tumors. It explores the potential clinical applications in tumor treatment, highlighting possible therapeutic targets and novel strategies for targeted tumor therapy.


Sujet(s)
Chimiokine CXCL12 , Tumeurs , Récepteurs CXCR4 , Facteur de transcription STAT-3 , Transduction du signal , Humains , Facteur de transcription STAT-3/métabolisme , Récepteurs CXCR4/métabolisme , Chimiokine CXCL12/métabolisme , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Animaux , Récepteurs CXCR/métabolisme , Récepteurs CXCR/génétique
3.
Cell Death Dis ; 15(6): 434, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898023

RÉSUMÉ

The interaction between glioblastoma cells and glioblastoma-associated macrophages (GAMs) influences the immunosuppressive tumor microenvironment, leading to ineffective immunotherapies. We hypothesized that disrupting the communication between tumors and macrophages would enhance the efficacy of immunotherapies. Transcriptomic analysis of recurrent glioblastoma specimens indicated an enhanced neuroinflammatory pathway, with CXCL12 emerging as the top-ranked gene in secretory molecules. Single-cell transcriptome profiling of naïve glioblastoma specimens revealed CXCL12 expression in tumor and myeloid clusters. An analysis of public glioblastoma datasets has confirmed the association of CXCL12 with disease and PD-L1 expression. In vitro studies have demonstrated that exogenous CXCL12 induces pro-tumorigenic characteristics in macrophage-like cells and upregulated PD-L1 expression through NF-κB signaling. We identified CXCR7, an atypical receptor for CXCL12 predominantly present in tumor cells, as a negative regulator of CXCL12 expression by interfering with extracellular signal-regulated kinase activation. CXCR7 knockdown in a glioblastoma mouse model resulted in worse survival outcomes, increased PD-L1 expression in GAMs, and reduced CD8+ T-cell infiltration compared with the control group. Ex vivo T-cell experiments demonstrated enhanced cytotoxicity against tumor cells with a selective CXCR7 agonist, VUF11207, reversing GAM-induced immunosuppression in a glioblastoma cell-macrophage-T-cell co-culture system. Notably, VUF11207 prolonged survival and potentiated the anti-tumor effect of the anti-PD-L1 antibody in glioblastoma-bearing mice. This effect was mitigated by an anti-CD8ß antibody, indicating the synergistic effect of VUF11207. In conclusion, CXCL12 conferred immunosuppression mediated by pro-tumorigenic and PD-L1-expressing GAMs in glioblastoma. Targeted activation of glioblastoma-derived CXCR7 inhibits CXCL12, thereby eliciting anti-tumor immunity and enhancing the efficacy of anti-PD-L1 antibodies.


Sujet(s)
Antigène CD274 , Chimiokine CXCL12 , Glioblastome , Récepteurs CXCR , Glioblastome/anatomopathologie , Glioblastome/immunologie , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Animaux , Récepteurs CXCR/métabolisme , Récepteurs CXCR/génétique , Chimiokine CXCL12/métabolisme , Souris , Antigène CD274/métabolisme , Lignée cellulaire tumorale , Microenvironnement tumoral , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
4.
Nat Commun ; 15(1): 4611, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38816420

RÉSUMÉ

G protein-coupled receptors (GPCRs) are pivotal therapeutic targets, but their complex structure poses challenges for effective drug design. Nanobodies, or single-domain antibodies, have emerged as a promising therapeutic strategy to target GPCRs, offering advantages over traditional small molecules and antibodies. However, an incomplete understanding of the structural features enabling GPCR-nanobody interactions has limited their development. In this study, we investigate VUN701, a nanobody antagonist targeting the atypical chemokine receptor 3 (ACKR3). We determine that an extended CDR3 loop is required for ACKR3 binding. Uncommon in most nanobodies, an extended CDR3 is prevalent in GPCR-targeting nanobodies. Combining experimental and computational approaches, we map an inhibitory ACKR3-VUN701 interface and define a distinct conformational mechanism for GPCR inactivation. Our results provide insights into class A GPCR-nanobody selectivity and suggest a strategy for the development of these new therapeutic tools.


Sujet(s)
Récepteurs CXCR , Anticorps à domaine unique , Anticorps à domaine unique/composition chimique , Anticorps à domaine unique/métabolisme , Humains , Récepteurs CXCR/métabolisme , Récepteurs CXCR/génétique , Récepteurs CXCR/antagonistes et inhibiteurs , Récepteurs CXCR/composition chimique , Cellules HEK293 , Liaison aux protéines , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/composition chimique , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Animaux
5.
Int J Mol Sci ; 25(9)2024 May 04.
Article de Anglais | MEDLINE | ID: mdl-38732237

RÉSUMÉ

NanoLuc-mediated bioluminescence resonance energy transfer (NanoBRET) has gained popularity for its ability to homogenously measure ligand binding to G protein-coupled receptors (GPCRs), including the subfamily of chemokine receptors. These receptors, such as ACKR3, CXCR4, CXCR3, play a crucial role in the regulation of the immune system, are associated with inflammatory diseases and cancer, and are seen as promising drug targets. The aim of this study was to optimize NanoBRET-based ligand binding to NLuc-ACKR3 and NLuc-CXCR4 using different fluorescently labeled chemokine CXCL12 analogs and their use in a multiplex NanoBRET binding assay of two chemokine receptors at the same time. The four fluorescent CXCL12 analogs (CXCL12-AZD488, -AZD546, -AZD594, -AZD647) showed high-affinity saturable binding to both NLuc-ACKR3 and NLuc-CXCR4, with relatively low levels of non-specific binding. Additionally, the binding of all AZDye-labeled CXCL12s to Nluc receptors was inhibited by pharmacologically relevant unlabeled chemokines and small molecules. The NanoBRET binding assay for CXCL10-AZD488 binding to Nluc-CXCR3 was also successfully established and successfully employed for the simultaneous measurement of the binding of unlabeled small molecules to NLuc-CXCR3 and NLuc-CXCR4. In conclusion, multiplexing the NanoBRET-based competition binding assay is a promising tool for testing unlabeled (small) molecules against multiple GPCRs simultaneously.


Sujet(s)
Chimiokine CXCL12 , Liaison aux protéines , Récepteurs CXCR3 , Récepteurs CXCR4 , Récepteurs CXCR , Humains , Récepteurs CXCR4/métabolisme , Récepteurs CXCR/métabolisme , Récepteurs CXCR/génétique , Chimiokine CXCL12/métabolisme , Récepteurs CXCR3/métabolisme , Techniques de transfert d'énergie par résonance de bioluminescence/méthodes , Ligands , Colorants fluorescents/composition chimique
6.
Development ; 151(4)2024 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-38300826

RÉSUMÉ

ACKR3 scavenges and degrades the stem cell recruiting chemokine CXCL12, which is essential for proper embryonic and, in particular, haematopoietic development. Here, we demonstrate strong expression of ACKR3 on trophoblasts. Using a maternally administered pharmacological blocker and Cre-mediated genetic approaches, we demonstrate that trophoblast ACKR3 is essential for preventing movement of CXCL12 from the mother to the embryo, with elevated plasma CXCL12 levels being detected in embryos from ACKR3-blocker-treated mothers. Mice born to mothers treated with the blocker are lighter and shorter than those born to vehicle-treated mothers and, in addition, display profound anaemia associated with a markedly reduced bone marrow haematopoietic stem cell population. Importantly, although the haematopoietic abnormalities are corrected as mice age, our studies reveal a postnatal window during which offspring of ACKR3-blocker-treated mice are unable to mount effective inflammatory responses to inflammatory/infectious stimuli. Overall, these data demonstrate that ACKR3 is essential for preventing CXCL12 transfer from mother to embryo and for ensuring properly regulated CXCL12 control over the development of the haematopoietic system.


Sujet(s)
Placenta , Récepteurs CXCR , Animaux , Femelle , Souris , Grossesse , Chimiokine CXCL12/génétique , Chimiokine CXCL12/métabolisme , Mouvement , Mutation , Placenta/métabolisme , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Transduction du signal/génétique
7.
Ren Fail ; 46(1): 2300727, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38189094

RÉSUMÉ

Renal fibrosis is a common feature of various chronic kidney diseases. However, the underlying mechanism remains poorly understood. The CXC chemokine receptor (CXCR) family plays a role in renal fibrosis; however, the detailed mechanisms have not been elucidated. In this study, we investigated the potential role of CXCR7 in mediating renal fibrosis. CXCR7 expression is decreased in unilateral ischemia-reperfusion injury (UIRI) and unilateral ureteral obstruction mouse models. Furthermore, CXCR7 was specifically expressed primarily in the Lotus Tetragonolobus Lectin-expressing segment of tubules, was slightly expressed in the peanut agglutinin-expressing segment, and was barely expressed in the Dolichos biflorus agglutinin-expressing segment. Administration of pFlag-CXCR7, an overexpression plasmid for CXCR7, significantly inhibited the activation of ß-catenin signaling and protected against the progression of epithelial-to-mesenchymal transition (EMT) and renal fibrosis in a UIRI mouse model. Using cultured HKC-8 cells, we found that CXCR7 significantly downregulated the expression of active ß-catenin and fibrosis-related markers, including fibronectin, Collagen I, and α-SMA. Furthermore, CXCR7 significantly attenuated TGF-ß1-induced changes in ß-catenin signaling, EMT and fibrosis. These results suggest that CXCR7 plays a crucial role in inhibiting the activation of ß-catenin signaling and the progression of EMT and renal fibrosis. Thus, CXCR7 could be a novel therapeutic target for renal fibrosis.


Sujet(s)
Maladies du rein , Récepteurs CXCR , Animaux , Souris , bêta-Caténine , Modèles animaux de maladie humaine , Cellules épithéliales , Transition épithélio-mésenchymateuse , Fibrose , Maladies du rein/étiologie , Récepteurs CXCR/génétique
8.
Oncol Rep ; 51(2)2024 02.
Article de Anglais | MEDLINE | ID: mdl-38099418

RÉSUMÉ

C­X­C motif chemokine 12 (CXCL12) promotes metastasis of several tumors by affecting cell migration and invasion via its receptors, C­X­C chemokine receptor type (CXCR)4 and CXCR7. Current therapeutic approaches focus on the selective inactivation of either CXCR4 or CXCR7 in patients with cancer. Alternative strategies may emerge from the analysis of downstream events that mediate the migratory effects of CXCL12 in cancer cells. While CXCR4 activates cell signaling through both G proteins and arrestins, CXCR7 is believed to preferentially signal through arrestins. The present study analyzed the CXCL12­dependent chemotaxis of A549, C33A, DLD­1, MDA­MB­231 and PC­3 cells, in which either the activity of G proteins, EGFR or Src kinase was inhibited pharmacologically or the expression of arrestins was inhibited by RNA interference. The results demonstrated that CXCL12­induced migration of A549, C33A, DLD­1, MDA­MB­231 and PC­3 cells was attenuated by the Gαi/o­inhibitor pertussis toxin (PTX), but was unaffected by small interfering RNA­mediated gene silencing of ß­arrestin1/2. In particular, the sensitivity of DLD­1 migration to PTX was unexpected, as it is solely dependent on the non­classical chemokine receptor, CXCR7. Furthermore, chemotactic responses to CXCL12 were additionally prevented by inhibiting EGFR activity via AG1478 and Src kinase activity via Src inhibitor­1. In conclusion, the results of the present study suggest that G protein­ and Src­dependent transactivation of EGFR is a common mechanism through which CXCL12­bound CXCR4 and/or CXCR7 control cancer cell migration and metastasis. These findings highlight EGFR as a potential therapeutic target that interferes with CXCL12­induced cancer expansion.


Sujet(s)
Tumeurs , Récepteurs CXCR , Humains , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Activation de la transcription , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Transduction du signal , Protéines G , Chimiokine CXCL12/génétique , Chimiokine CXCL12/métabolisme , Mouvement cellulaire , Arrestines/génétique , Arrestines/métabolisme , Arrestines/pharmacologie , src-Family kinases/génétique , src-Family kinases/métabolisme , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme
9.
Nat Commun ; 14(1): 4808, 2023 08 09.
Article de Anglais | MEDLINE | ID: mdl-37558722

RÉSUMÉ

Chemokine receptors constitute an important subfamily of G protein-coupled receptors (GPCRs), and they are critically involved in a broad range of immune response mechanisms. Ligand promiscuity among these receptors makes them an interesting target to explore multiple aspects of biased agonism. Here, we comprehensively characterize two chemokine receptors namely, CXCR4 and CXCR7, in terms of their transducer-coupling and downstream signaling upon their stimulation by a common chemokine agonist, CXCL12, and a small molecule agonist, VUF11207. We observe that CXCR7 lacks G-protein-coupling while maintaining robust ßarr recruitment with a major contribution of GRK5/6. On the other hand, CXCR4 displays robust G-protein activation as expected but exhibits significantly reduced ßarr-coupling compared to CXCR7. These two receptors induce distinct ßarr conformations even when activated by the same agonist, and CXCR7, unlike CXCR4, fails to activate ERK1/2 MAP kinase. We also identify a key contribution of a single phosphorylation site in CXCR7 for ßarr recruitment and endosomal localization. Our study provides molecular insights into intrinsic-bias encoded in the CXCR4-CXCR7 system with broad implications for drug discovery.


Sujet(s)
Récepteurs CXCR , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Récepteurs CXCR4/métabolisme , Transduction du signal , Protéines G , Mitogen-Activated Protein Kinase 3/métabolisme , Chimiokine CXCL12/métabolisme
10.
FEBS Lett ; 597(15): 2017-2027, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37395117

RÉSUMÉ

It is unknown whether heteromerization between chemokine (C-X-C motif) receptor 4 (CXCR4), atypical chemokine receptor 3 (ACKR3) and α1b -adrenoceptor (α1b -AR) influences effects of the CXCR4/ACKR3 agonist chemokine (C-X-C motif) ligand 12 (CXCL12) and the noncognate CXCR4 agonist ubiquitin on agonist-promoted G protein activation. We provide biophysical evidence that both ligands stimulate CXCR4-mediated Gαi activation. Unlike CXCL12, ubiquitin fails to recruit ß-arrestin. Both ligands differentially modulate the conformation of CXCR4:ACKR3 heterodimers and its propensity to hetero-trimerize with α1b -AR. CXCR4:ACKR3 heterodimerization reduces the potency of CXCL12, but not of ubiquitin, to activate Gαi. Ubiquitin enhances phenylephrine-stimulated α1b -AR-promoted Gαq activation from hetero-oligomers comprising CXCR4. CXCL12 enhances phenylephrine-stimulated α1b -AR-promoted Gαq activation from CXCR4:α1b -AR heterodimers and reduces phenylephrine-stimulated α1b -AR-promoted Gαq activation from ACKR3 comprising heterodimers and trimers. Our findings suggest heteromer and ligand-dependent functions of the receptor partners.


Sujet(s)
Récepteurs CXCR4 , Récepteurs CXCR , Ligands , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Chimiokine CXCL12/métabolisme , Phényléphrine/pharmacologie , Protéines G/métabolisme , Ubiquitine/métabolisme , Récepteurs adrénergiques/métabolisme
11.
Zhongguo Gu Shang ; 36(6): 514-8, 2023 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-37366092

RÉSUMÉ

OBJECTIVE: To investigate the enhancement of macrophage chemotaxis in patients with knee osteoarthritis (KOA) and its correlation with the disease severity. METHODS: Eighty patients with KOA admitted from July 2019 to June 2022 were enrolled as the observation group and divided into 29 cases of moderate group, 30 cases of severe group and 21 cases of extremely severe group. At the same time, 30 healthy subjects were included as the control group. The gene expressions of NF-κB, CXC chemokine receptor 7 (CXCR7) and CXC chemokine ligand 12 (CXCL12) in macrophages of each group were analyzed. Visual analogue scale(VAS) was used to evaluate the degree of joint pain. Joint function was evaluated by knee Joint Society Scoring system(KSS). Finally, data analysis was carried out. RESULTS: The expression levels of NF-κB, CXCR7 and CXCL12 in moderate group, severe group and extreme recombination group were higher than those in control group. The VAS, the expression of NF-κB, CXCR7 and CXCL12 in the severe group and the extreme recombination group were higher than those in the moderate group, whereas KSS was lower than that in the moderate group. The VAS, expression levels of NF-κB, CXCR7 and CXCL12 in the extremely severe group were higher than those in the severe group, and KSS was lower than that in the severe group (all P<0.01). The expression levels of NF-κB, CXCR7 and CXCL12 in macrophages were positively correlated with VAS score, but negatively correlated with KSS(all P<0.01). The expression levels of NF-κB, CXCR7 and CXCL12 in macrophages were positively correlated with the severity of disease. After excluding the influence of traditional factors (gender, age and disease duration), multiple linear regression analysis further showed that the expression levels of NF-κB, CXCR7 and CXCL12 were still positively correlated with the severity of disease(all P<0.01). CONCLUSION: The chemotaxis of macrophages in patients with KOA increased with the aggravation of the disease, and was related to the degree of pain and function impairment.


Sujet(s)
Gonarthrose , Récepteurs CXCR , Humains , Gonarthrose/génétique , Chimiotaxie/génétique , Facteur de transcription NF-kappa B/génétique , Facteur de transcription NF-kappa B/métabolisme , Macrophages/métabolisme , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Acuité des besoins du patient
12.
J Clin Invest ; 133(15)2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37347559

RÉSUMÉ

CXCR7 is an atypical chemokine receptor that recruits ß-arrestin (ARRB2) and internalizes into clathrin-coated intracellular vesicles where the complex acts as a scaffold for cytoplasmic kinase assembly and signal transduction. Here, we report that CXCR7 was elevated in the majority of prostate cancer (PCa) cases with neuroendocrine features (NEPC). CXCR7 markedly induced mitotic spindle and cell cycle gene expression. Mechanistically, we identified Aurora Kinase A (AURKA), a key regulator of mitosis, as a novel target that was bound and activated by the CXCR7-ARRB2 complex. CXCR7 interacted with proteins associated with microtubules and golgi, and, as such, the CXCR7-ARRB2-containing vesicles trafficked along the microtubules to the pericentrosomal golgi apparatus, where the complex interacted with AURKA. Accordingly, CXCR7 promoted PCa cell proliferation and tumor growth, which was mitigated by AURKA inhibition. In summary, our study reveals a critical role of CXCR7-ARRB2 in interacting and activating AURKA, which can be targeted by AURKA inhibitors to benefit a subset of patients with NEPC.


Sujet(s)
Tumeurs de la prostate , Récepteurs CXCR , Mâle , Humains , Aurora kinase A/génétique , Aurora kinase A/métabolisme , Transduction du signal , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Tumeurs de la prostate/anatomopathologie , Prolifération cellulaire , Lignée cellulaire tumorale
13.
PLoS One ; 18(5): e0285597, 2023.
Article de Anglais | MEDLINE | ID: mdl-37252916

RÉSUMÉ

Atypical chemokine receptor 3 (ACKR3) is a scavenger of the chemokines CXCL11 and CXCL12 and of several opioid peptides. Additional evidence indicates that ACKR3 binds two other non-chemokine ligands, namely the peptide hormone adrenomedullin (AM) and derivatives of the proadrenomedullin N-terminal 20 peptide (PAMP). AM exhibits multiple functions in the cardiovascular system and is essential for embryonic lymphangiogenesis in mice. Interestingly, AM-overexpressing and ACKR3-deficient mouse embryos both display lymphatic hyperplasia. Moreover, in vitro evidence suggested that lymphatic endothelial cells (LECs), which express ACKR3, scavenge AM and thereby reduce AM-induced lymphangiogenic responses. Together, these observations have led to the conclusion that ACKR3-mediated AM scavenging by LECs serves to prevent overshooting AM-induced lymphangiogenesis and lymphatic hyperplasia. Here, we further investigated AM scavenging by ACKR3 in HEK293 cells and in human primary dermal LECs obtained from three different sources in vitro. LECs efficiently bound and scavenged fluorescent CXCL12 or a CXCL11/12 chimeric chemokine in an ACKR3-dependent manner. Conversely, addition of AM induced LEC proliferation but AM internalization was found to be independent of ACKR3. Similarly, ectopic expression of ACKR3 in HEK293 cells did not result in AM internalization, but the latter was avidly induced upon co-transfecting HEK293 cells with the canonical AM receptors, consisting of calcitonin receptor-like receptor (CALCRL) and receptor activity-modifying protein (RAMP)2 or RAMP3. Together, these findings indicate that ACKR3-dependent scavenging of AM by human LECs does not occur at ligand concentrations sufficient to trigger AM-induced responses mediated by canonical AM receptors.


Sujet(s)
Adrénomédulline , Cellules endothéliales , Récepteurs CXCR , Humains , Adrénomédulline/génétique , Chimiokine CXCL11 , Cellules endothéliales/métabolisme , Cellules HEK293 , Hyperplasie , Récepteurs à l'adrénomédulline , Récepteurs CXCR/génétique
14.
Neuro Oncol ; 25(10): 1775-1787, 2023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37074930

RÉSUMÉ

BACKGROUND: Meningiomas are mainly benign brain tumors, although about 20% of histologically benign cases are clinically aggressive and recur after resection. We hypothesize that meningioma brain invasiveness and recurrence may be related to the presence of cancer stem cells and their high responsiveness to the CXCL12-CXCR4/CXCR7 chemokine axis. The aim of this study was to isolate meningioma stem cells from human samples, characterize them for biological features related to malignant behavior, and to identify the role of CXCR4/CXCR7 in these processes. METHODS: Meningioma stem cells were isolated from patient-derived primary cultures in stem cell-permissive conditions, and characterized for phenotype, self-renewal, proliferation and migration rates, vasculogenic mimicry (VM), and in vivo tumorigenesis, in comparison with differentiated meningioma cells and stem-like cells isolated from normal meninges. These cell populations were challenged with CXCL12 and CXCL11 and receptor antagonists to define the chemokine role in stem cell-related functions. RESULTS: Stem-like cells isolated from meningioma cultures display higher proliferation and migration rates, and VM, as compared to meningioma non-stem cells or cells isolated from normal meninges and were the only tumorigenic population in vivo. In meningioma cells, these stem-like functions were under the control of the CXCR4/CXCR7 chemokine axis. CONCLUSIONS: We report a role for CXCL11 and CXCL12 in the control of malignant features in stem-like cells isolated from human meningioma, providing a possible basis for the aggressive clinical behavior observed in subsets of these tumors. CXCR4/CXCR7 antagonists might represent a useful approach for meningioma at high risk of recurrence and malignant progression.


Sujet(s)
Tumeurs du cerveau , Tumeurs des méninges , Méningiome , Récepteurs CXCR , Humains , Chimiokine CXCL12/génétique , Récepteurs CXCR/génétique , Récepteurs CXCR4/génétique , Transduction du signal , Chimiokine CXCL11
15.
Anticancer Agents Med Chem ; 23(20): 2248-2253, 2023.
Article de Anglais | MEDLINE | ID: mdl-36748819

RÉSUMÉ

BACKGROUND: Desmoid tumors have an extremely variable natural history. The uncertainty behind desmoid behavior reflects the complexity, which subtends its development and non-linear advancement. Apart from Wnt- ßcatenin mutation, estrogen receptors, and COX-2 overexpression, little is known about the ability of desmoids to grow and recur while being unable to metastasize. Several tumors have been shown to express the CXCR4/CXCR7/CXCL12 axis, whose functions are essential for tumoral development. AIMS: This study aimed to investigate the expression of the CXCR4/CXCR7/CXCL12 axis in primary desmoid tumors and discuss the potential role of this key-signaling as an antiangiogenic therapeutic strategy. METHODS: In this study, 3 µm-thick consecutive sections from each formalin-fixed and paraffin-embedded tissue block were treated with mouse monoclonal antibodies developed against CD34, CXCR4, CXCR7, and CXCL12. RESULTS: Two distinct vessel populations: CXCR4+ and CXCR4- vessels, have been found. Similarly, chemokine receptor CXCR7 expression in the entire desmoid tumor series positively stained a portion of tumor-associated vessels, identifying two distinct subpopulations of vessels: CXCR7+ and CXCR7- vessels. All 8 neoplastic tissue samples expressed CXCL12. Immunohistochemical positivity was identified in both stromal and endothelial vascular cells. Compared to CXCR4 and CXCR7, the vast majority of tumor-associated vessels were found to express this chemokine. CONCLUSION: It is the first time, as per our knowledge, that CXCR4/CXCR7/CXCL12 axis expression has been identified in a desmoid type-fibromatosis series. CXCL12 expression by neoplastic cells, together with CXCR4 and CXCR7 expression by a subgroup of tumor-associated vessels, was detected in all desmoid tumor tissue samples examined. Since chemokines are known contributors to neovascularization, CXCR4/CXCR7/CXCL12 axis may play a role in angiogenesis in this soft-tissue tumor histotype, thereby supporting its growth.


Sujet(s)
Fibromatose agressive , Récepteurs CXCR , Animaux , Souris , Prolifération cellulaire , Récidive tumorale locale , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Transduction du signal , Chimiokine CXCL12/génétique , Chimiokine CXCL12/métabolisme , Récepteurs des oestrogènes
16.
CNS Neurosci Ther ; 29(2): 712-726, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36523152

RÉSUMÉ

AIMS: Chemokine receptor 7 (CXCR7) exerts protective effects on the brain. MicroRNAs (miRNAs) are involved in cerebral ischemia/reperfusion (I/R) injury, but their involvement in CXCR7-mediated brain protection is unknown. In this study, we investigated the role of miRNAs in CXCR7-mediated brain protection. METHODS: CXCR7 levels in peripheral blood samples from patients with acute ischemic stroke (AIS) and ischemic penumbra area brain tissues from middle cerebral artery occlusion (MCAO) rats after recanalization were measured. An miRNA microarray analysis was performed to examine the expression of miRNAs caused by CXCR7 knockdown in ischemic penumbra area brain tissue in middle cerebral artery occlusion-reperfusion rats and to predict corresponding downstream target genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed the most enriched pathways. A dual-luciferase reporter assay confirmed the direct regulation of miR-182 on the target gene TCF7L2. The correlation between TCF7L2 and CXCR7/miR-182 was verified using rescue assays. RESULTS: CXCR7 expression was upregulated in MCAO rats and mechanical thrombectomy patients with AIS compared to that in controls. The motor and sensory functions of MCAO rats with CXCR7 knockdown further decreased, and the infarct volume and cerebral edema increased. miRNA microarray data showed that seven miRNAs were differentially expressed after shRNA-CXCR7 treatment. The dual-luciferase reporter assay confirmed that miR-182 directly targeted the TCF7L2 gene. Rescue assays confirmed that TCF7L2 is downstream of CXCR7/miR-182. KEGG pathway analysis showed that the Hippo pathway may be a key pathway in CXCR7 upregulation and plays a role in protecting the brain after interventional surgery. Animal experiments have shown that CXCR7-mediated cerebral I/R injury promotes the phosphorylation of key molecules YAP and TAZ in the Hippo pathway. CONCLUSION: CXCR7 protects against cerebral I/R injury, possibly via the miR-182/TCF7L2/Hippo pathway. These results indicate that CXCR7 affects cerebral ischemia-reperfusion injury through miRNA regulation and downstream pathways.


Sujet(s)
Encéphalopathie ischémique , Accident vasculaire cérébral ischémique , microARN , Récepteurs CXCR , Lésion d'ischémie-reperfusion , Rats , Animaux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Rat Sprague-Dawley , Encéphalopathie ischémique/génétique , Encéphalopathie ischémique/métabolisme , microARN/génétique , microARN/métabolisme , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/métabolisme , Récepteurs CXCR/génétique
17.
BMC Cancer ; 22(1): 1335, 2022 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-36539774

RÉSUMÉ

BACKGROUND: The chemokines, CXCL12 and CXCL11, are upregulated in tumors from many organs and control their progression. CXCL12 and CXCL11 affect tumor cell functions by either binding their prime receptors, CXCR4 and CXCR3, respectively, and/or CXCR7 as a common second chemokine receptor. In humans, CXCR3 exists in the functional splice variants, CXCR3A and CXCR3B, which either have pro- or anti-tumor activity, respectively. Despite the intimate crosstalk between the CXCL12- and CXCL11-system, the impact of a combination of CXCL12 and CXCL11 on tumor progression remains vague. METHODS: In the present work, we have analyzed CXCL12 and CXCL11 for combined effects on migration, invasion, proliferation, and cytostatic-induced apoptosis of the human tumor cells, A549, A767, A772, DLD-1, and MDA-MB-231. RESULTS: We demonstrate that the mode of interaction differs with respect to cell type and function and allows for either potentiation, attenuation or no changes of cellular responses. The divergent responses are not the result of the distinct use of different CXCL12- and CXCL11-receptors by the respective tumor cells, but in case of cell migration seem to be associated with the activation of p38 signaling pathways. CONCLUSIONS: Our findings point to therapeutic limitations of ongoing efforts to selectively target CXCR3, CXCR4, or CXCR7 in cancer patients, and rather favor individualized targeting strategies.


Sujet(s)
Tumeurs , Récepteurs CXCR , Humains , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Tumeurs/génétique , Récepteurs CXCR4/génétique , Récepteurs CXCR4/métabolisme , Chimiokine CXCL12/métabolisme , Transduction du signal , Mouvement cellulaire , Apoptose , Chimiokine CXCL11/génétique , Chimiokine CXCL11/métabolisme
18.
Int J Mol Sci ; 23(17)2022 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-36077241

RÉSUMÉ

CXCR7 and CXCR4 are G protein-coupled receptors (GPCRs) that can be stimulated by CXCL12 in various human cancers. CXCR7/4-CXCL12 binding can initiate activation of multiple pathways including JAK/STAT and manganese superoxide dismutase (MnSOD) signaling, and initiate epithelial-mesenchymal transition (EMT) process. It is established that cancer cell invasion and migration are caused because of these events. In particular, the EMT process is an important process that can determine the prognosis for cancer. Since the antitumor effect of leelamine (LEE) has been reported in various previous studies, here, we have evaluated the influence of LEE on the CXCR7/4 signaling axis and EMT processes. We first found that LEE suppressed expression of CXCR7 and CXCR4 both at the protein and mRNA levels, and showed inhibitory effects on these chemokines even after stimulation by CXCL12 ligand. In addition, LEE also reduced the level of MnSOD and inhibited the EMT process to attenuate the invasion and migration of breast cancer cells. In addition, phosphorylation of the JAK/STAT pathway, which acts down-stream of these chemokines, was also abrogated by LEE. It was also confirmed that LEE can induce an imbalance of GSH/GSSG and increases ROS, thereby resulting in antitumor activity. Thus, we establish that targeting CXCR7/4 in breast cancer cells can not only inhibit the invasion and migration of cancer cells but also can affect JAK/STAT, EMT process, and production of ROS. Overall, the findings suggest that LEE can function as a novel agent affecting the breast cancer.


Sujet(s)
Tumeurs du sein , Récepteurs CXCR , Abiétanes , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Chimiokine CXCL12/génétique , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/pharmacologie , Transition épithélio-mésenchymateuse , Femelle , Humains , Janus kinases/métabolisme , Espèces réactives de l'oxygène/métabolisme , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Récepteurs CXCR4/métabolisme , Facteurs de transcription STAT/métabolisme , Transduction du signal
19.
World J Surg Oncol ; 20(1): 259, 2022 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-35978426

RÉSUMÉ

BACKGROUND: CXC chemokine receptor gene family consists of seven well-established members which are broadly involved in biological functions of various cancers. Currently, limited studies have shed light on the expression profile of CXCR family members (CXCRs), as well as their prognostic value, in head and neck squamous cells carcinoma (HNSCC). METHODS: The data for this study were retrieved from the Cancer Genome Atlas database and other publicly available databases, including gene expression, methylation profiles, clinical information, immunological features, and prognoses. The expression pattern and prognostic values of CXCRs were identified, and the potential mechanism underlying CXCRs function in HNSCC was investigated by gene set enrichment analysis (GSEA). RESULTS: CXCRs were differentially expressed in HNSCC. As shown by Kaplan-Meier analysis, high CXCR3-6 expression was significantly associated with better prognostic outcomes of HNSCC patients, including overall survival and progression-free survival. According to the results of univariate and multivariate Cox proportional risk regression analysis, it was demonstrated that upregulation of CXCR3-6 was an independent factor for better prognosis, while the two other clinical features, age and stage, were factors for worse prognosis. A significant positive correlation between CXCR3-6 and tumor-infiltrated immune cells was revealed by results from Tumor Immune Estimation Resource and CIBERSORT analysis database. The main involvement of CXCRs in immune and inflammatory responses was further confirmed by GSEA. CONCLUSIONS: Overall, this study provided a rationale for targeting CXCRs as a promising therapeutic strategy of HNSCC.


Sujet(s)
Tumeurs de la tête et du cou , Récepteurs CXCR , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs de la tête et du cou/génétique , Humains , Estimation de Kaplan-Meier , Pronostic , Récepteurs CXCR/génétique , Récepteurs CXCR/métabolisme , Carcinome épidermoïde de la tête et du cou
20.
Science ; 377(6602): 222-228, 2022 07 08.
Article de Anglais | MEDLINE | ID: mdl-35857540

RÉSUMÉ

G protein-coupled receptors (GPCRs) recruit ß-arrestins to coordinate diverse cellular processes, but the structural dynamics driving this process are poorly understood. Atypical chemokine receptors (ACKRs) are intrinsically biased GPCRs that engage ß-arrestins but not G proteins, making them a model system for investigating the structural basis of ß-arrestin recruitment. Here, we performed nuclear magnetic resonance (NMR) experiments on 13CH3-ε-methionine-labeled ACKR3, revealing that ß-arrestin recruitment is associated with conformational exchange at key regions of the extracellular ligand-binding pocket and intracellular ß-arrestin-coupling region. NMR studies of ACKR3 mutants defective in ß-arrestin recruitment identified an allosteric hub in the receptor core that coordinates transitions among heterogeneously populated and selected conformational states. Our data suggest that conformational selection guides ß-arrestin recruitment by tuning receptor dynamics at intracellular and extracellular regions.


Sujet(s)
Récepteurs CXCR , bêta-Arrestines , Régulation allostérique , Ligands , Spectroscopie par résonance magnétique , Mutation , Liaison aux protéines , Conformation des protéines , Récepteurs CXCR/composition chimique , Récepteurs CXCR/génétique , bêta-Arrestines/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE