Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 579
Filtrar
1.
Development ; 151(4)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38300826

RESUMEN

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.


Asunto(s)
Placenta , Receptores CXCR , Animales , Femenino , Ratones , Embarazo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Movimiento , Mutación , Placenta/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Transducción de Señal/genética
2.
Oncol Rep ; 51(2)2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38099418

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores CXCR , Humanos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Activación Transcripcional , Receptores CXCR/genética , Receptores CXCR/metabolismo , Transducción de Señal , Proteínas de Unión al GTP , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Movimiento Celular , Arrestinas/genética , Arrestinas/metabolismo , Arrestinas/farmacología , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo
3.
Nat Commun ; 14(1): 4808, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37558722

RESUMEN

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.


Asunto(s)
Receptores CXCR , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Proteínas de Unión al GTP , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quimiocina CXCL12/metabolismo
4.
FEBS Lett ; 597(15): 2017-2027, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37395117

RESUMEN

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.


Asunto(s)
Receptores CXCR4 , Receptores CXCR , Ligandos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Quimiocina CXCL12/metabolismo , Fenilefrina/farmacología , Proteínas de Unión al GTP/metabolismo , Ubiquitina/metabolismo , Receptores Adrenérgicos/metabolismo
5.
J Clin Invest ; 133(15)2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37347559

RESUMEN

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.


Asunto(s)
Neoplasias de la Próstata , Receptores CXCR , Masculino , Humanos , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Transducción de Señal , Receptores CXCR/genética , Receptores CXCR/metabolismo , Neoplasias de la Próstata/patología , Proliferación Celular , Línea Celular Tumoral
6.
Zhongguo Gu Shang ; 36(6): 514-8, 2023 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-37366092

RESUMEN

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.


Asunto(s)
Osteoartritis de la Rodilla , Receptores CXCR , Humanos , Osteoartritis de la Rodilla/genética , Quimiotaxis/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Macrófagos/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Gravedad del Paciente
7.
PLoS One ; 18(2): e0280001, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36800350

RESUMEN

The chemokine SDF-1 (CXCL12) and its receptor CXCR4 control several processes during embryonic development such as the regulation of stem cell proliferation, differentiation, and migration. However, the role of this pathway in the formation of the pituitary gland is not understood. We sought to characterise the expression patterns of CXCR4, SDF-1 and CXCR7 at different stages of pituitary gland development. Our expression profiling revealed that SDF-1 is expressed in progenitor-rich regions of the pituitary anterior lobe, that CXCR4 and CXCR7 have opposite expression domains and that CXCR4 expression is conserved between mice and human embryos. We then assessed the importance of this signalling pathway in the development and function of the murine pituitary gland through conditional deletion of CXCR4 in embryonic pituitary progenitors. Successful and specific ablation of CXCR4 expression in embryonic pituitary progenitors did not lead to observable embryonic nor postnatal defects but allowed the identification of stromal CXCR4+ cells not derived from HESX1+ progenitors. Further analysis of constitutive SDF-1, CXCR7 and CXCR4 mutants of the pathway indicates that CXCR4 expression in HESX1+ cells and their descendants is not essential for normal pituitary development in mice.


Asunto(s)
Receptores CXCR , Animales , Femenino , Humanos , Ratones , Embarazo , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas de Homeodominio/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal
8.
Oncogene ; 42(11): 785-792, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36755058

RESUMEN

Chemokines and their cognate receptors comprise an intricate signaling network that becomes high-jacked by cancer cells for uncontrollable tumor growth and dissemination. ACKR3 (Atypical Chemokine Receptor 3), traditionally called CXCR7, is up-regulated in many cancers, including advanced prostate cancer, and represents promising targets for therapeutic intervention. Unlike typical G protein-coupled receptors such as CXCR4, CXCR7, once bound by its cognate ligand CXCL12, initiates the recruitment of ß-arrestin instead of G proteins, and results in rapid internalization and degradation of CXCL12, functioning as a scavenger receptor. However, recent evidence suggests that CXCR7 may be more than a scavenger or auxiliary receptor of CXCR4 and that it may play essential roles in regulating cancer progression, some of which are independent of CXCR4 and its ligands, such as CXCL12. Constitutively active CXCR7 binds to ß-arrestin. This protein complex internalizes to form a scaffold for assembling and activating various cytoplasmic kinases necessary for cell survival and tumor growth. Here we review and discuss the up-to-date knowledge on CXCR7 regulation and function and how this new understanding guides the development of CXCR7 inhibitors, focusing on prostate cancer.


Asunto(s)
Neoplasias de la Próstata , Receptores CXCR , Humanos , Masculino , beta-Arrestinas/metabolismo , Quimiocina CXCL12/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología
9.
Anticancer Agents Med Chem ; 23(20): 2248-2253, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36748819

RESUMEN

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.


Asunto(s)
Fibromatosis Agresiva , Receptores CXCR , Animales , Ratones , Proliferación Celular , Recurrencia Local de Neoplasia , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Receptores de Estrógenos
10.
BMC Cancer ; 22(1): 1335, 2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36539774

RESUMEN

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.


Asunto(s)
Neoplasias , Receptores CXCR , Humanos , Receptores CXCR/genética , Receptores CXCR/metabolismo , Neoplasias/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Quimiocina CXCL12/metabolismo , Transducción de Señal , Movimiento Celular , Apoptosis , Quimiocina CXCL11/genética , Quimiocina CXCL11/metabolismo
11.
Int J Mol Sci ; 23(19)2022 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-36233192

RESUMEN

A better understanding of the complex crosstalk among key receptors and signaling pathways involved in cancer progression is needed to improve current therapies. We have investigated in cell models representative of the major subtypes of breast cancer (BC) the interplay between the chemokine CXCL12/CXCR4/ACKR3 and EGF receptor (EGFR) family signaling cascades. These cell lines display a high heterogeneity in expression profiles of CXCR4/ACKR3 chemokine receptors, with a predominant intracellular localization and different proportions of cell surface CXCR4+, ACKR3+ or double-positive cell subpopulations, and display an overall modest activation of oncogenic pathways in response to exogenous CXCL12 alone. Interestingly, we find that in MDA-MB-361 (luminal B subtype, Her2-overexpressing), but not in MCF7 (luminal A) or MDA-MB-231 (triple negative) cells, CXCR4/ACKR3 and EGFR receptor families share signaling components and crosstalk mechanisms to concurrently promote ERK1/2 activation, with a key involvement of the G protein-coupled receptor kinase 2 (GRK2) signaling hub and the cytosolic tyrosine kinase Src. Our findings suggest that in certain BC subtypes, a relevant cooperation between CXCR4/ACKR3 and growth factor receptors takes place to integrate concurrent signals emanating from the tumor microenvironment and foster cancer progression.


Asunto(s)
Neoplasias de la Mama , Receptores CXCR4 , Receptores CXCR , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Quimiocina CXCL12/metabolismo , Receptores ErbB/metabolismo , Femenino , Humanos , Proteínas Tirosina Quinasas/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transducción de Señal , Microambiente Tumoral
12.
J Med Chem ; 65(19): 13365-13384, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36150079

RESUMEN

The atypical chemokine receptor 3 (ACKR3), formerly known as CXC-chemokine receptor 7 (CXCR7), has been postulated to regulate platelet function and thrombus formation. Herein, we report the discovery and development of first-in-class ACKR3 agonists, which demonstrated superagonistic properties with Emax values of up to 160% compared to the endogenous reference ligand CXCL12 in a ß-arrestin recruitment assay. Initial in silico screening using an ACKR3 homology model identified two hits, C10 (EC50 19.1 µM) and C11 (EC50 = 11.4 µM). Based on these hits, extensive structure-activity relationship studies were conducted by synthesis and testing of derivatives. It resulted in the identification of the novel thiadiazolopyrimidinone-based compounds 26 (LN5972, EC50 = 3.4 µM) and 27 (LN6023, EC50 = 3.5 µM). These compounds are selective for ACKR3 versus CXCR4 and show metabolic stability. In a platelet degranulation assay, these agonists effectively reduced P-selectin expression by up to 97%, suggesting potential candidates for the treatment of platelet-mediated thrombosis.


Asunto(s)
Selectina-P , Receptores CXCR , Arrestina/metabolismo , Quimiocina CXCL12/metabolismo , Ligandos , Selectina-P/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología , beta-Arrestinas/metabolismo
13.
Int J Mol Sci ; 23(17)2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36077241

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Receptores CXCR , Abietanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , Transición Epitelial-Mesenquimal , Femenino , Humanos , Quinasas Janus/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
14.
Mol Immunol ; 151: 204-217, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36179603

RESUMEN

Atypical chemokine receptor 3 (ACKR3) has been linked to the tumor microenvironment. This work investigates the effects of ACKR3 and its regulatory molecules on the chemotactic migration of tumor-associated macrophages (TAMs) in hepatocellular carcinoma (HCC). RT-qPCR and western blot assays identified elevated ACKR3 and HDAC2 levels in HCC tissues and cells. Knockdown or overexpression of ACKR3 was induced in HCC cells through vectors of lentivirus plasmids, and then the conditioned medium of the HCC cells was collected to stimulate TAMs. The stimulated TAMs were co-cultured with CD3+ T cells. ACKR3 knockdown in HCC cells inhibited migration of TAMs and promoted M1 polarization, which restored the activity of T cells. Histone deacetylase 2 (HDAC2) recruited signal transducer and activator of transcription 1 (STAT1) to the ACKR3 promoter to activate ACKR3 transcription. HDAC2 silencing suppressed nuclear translocation of STAT1 and decreased ACKR3 expression. HDAC2 knockdown in HCC cells similarly suppressed TAM migration, promoted M1 polarization, and restored T cell function, but these changes were inversed by ACKR3 upregulation. HDAC2 or ACKR3 silencing weakened tumor growth and immune escape in mice. In conclusion, this study demonstrates that HDAC2 upregulates ACKR3 via STAT1 to induce migration of M2 macrophages and immune escape in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores CXCR/metabolismo , Animales , Línea Celular Tumoral , Medios de Cultivo Condicionados/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Macrófagos , Ratones , Factor de Transcripción STAT1/metabolismo , Microambiente Tumoral , Regulación hacia Arriba
15.
World J Surg Oncol ; 20(1): 259, 2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35978426

RESUMEN

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.


Asunto(s)
Neoplasias de Cabeza y Cuello , Receptores CXCR , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Estimación de Kaplan-Meier , Pronóstico , Receptores CXCR/genética , Receptores CXCR/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello
16.
J Biol Chem ; 298(10): 102382, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35973511

RESUMEN

Class A tick evasins are natural chemokine-binding proteins that block the signaling of multiple chemokines from the CC subfamily through their cognate receptors, thus suppressing leukocyte recruitment and inflammation. Development of tick evasins as chemokine-targeted anti-inflammatory therapeutics requires an understanding of the factors controlling their chemokine recognition and selectivity. To investigate the role of the evasin N-terminal region for chemokine recognition, we prepared chimeric evasins by interchanging the N-terminal regions of four class A evasins, including a newly identified evasin, EVA-RPU02. We show through chemokine binding analysis of the parental and chimeric evasins that the N-terminal region is critical for chemokine binding affinity and selectivity. Notably, we found some chimeras were unable to bind certain cognate chemokine ligands of both parental evasins. Moreover, unlike any natural evasins characterized to date, some chimeras exhibited specific binding to a single chemokine. These results indicate that the evasin N terminus interacts cooperatively with the "body" of the evasin to enable optimum chemokine recognition. Furthermore, the altered chemokine selectivity of the chimeras validates the approach of engineering the N termini of evasins to yield unique chemokine recognition profiles.


Asunto(s)
Proteínas de Artrópodos , Quimiocinas , Receptores CXCR , Rhipicephalus , Proteínas y Péptidos Salivales , Animales , Proteínas de Artrópodos/metabolismo , Quimiocinas/metabolismo , Unión Proteica , Receptores CXCR/metabolismo , Rhipicephalus/metabolismo , Transducción de Señal , Proteínas y Péptidos Salivales/metabolismo
17.
Shock ; 58(1): 56-67, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35984761

RESUMEN

ABSTRACT: Objectives: Nucleus pulposus (NP) cell degeneration promotes the progression of intervertebral disc (IVD) degeneration. MicroRNAs (miRs) are associated with IVD degeneration. This study expounded the mechanism of microRNA (miR)-25-3p carried by extracellular vesicles (EVs) derived from platelet-rich plasma (PRP) in interleukin (IL)-1ß-induced NP cell degeneration. Methods: Platelet-rich plasma from mouse blood was obtained, and EVs were isolated from PRP (EVs derived from PRP [PRP-EVs]) and identified. Nucleus pulposus cells were isolated from the mouse lumbar IVD and treated with IL-1ß to induce NP cell degeneration. Extracellular vesicles derived from PRP were added into NP cell culture medium. Afterward, intracellular miR-25-3p, sex determining region Y-related high-mobility-group box 4 (SOX4), and CXC chemokine receptor 7 (CXCR7) levels were examined. Nucleus pulposus cell viability, apoptosis, and inflammation were detected. Extracellular vesicles derived from PRP were labeled by PKH67 to obverse the uptake of EVs by NP cells. The binding relations between SOX4 and miR-25-3p and CXCR7 were predicted and examined. Functional rescue experiments were performed to investigate the roles of miR-25-3p, SOX4, and CXCR7 in NP cell degeneration. Results: miR-25-3p was downregulated, whereas SOX4 and CXCR7 were upregulated in IL-1ß-induced NP cells. Extracellular vesicles derived from PRP increased the cell viability, and decreased apoptosis and inflammation. miR-25-3p carried by PRP-EVs into NP cells alleviated NP cell degeneration. miR-25-3p inhibited SOX4 expression and limited CXCR7 transcription. Silencing miR-25-3p or overexpressing SOX4 or CXCR7 reversed the alleviating role of PRP-EVs in NP cell degeneration. Conclusion: miR-25-3p carried by PRP-EVs into NP cells elevated intracellular miR-25-3p expression, which suppressed SOX4 expression and further limited CXCR7 transcription, thus alleviating IL-1ß-induced NP cell degeneration. Extracellular vesicles derived from PRP containing miR-25-3p may be a new method for IVD treatment.


Asunto(s)
Vesículas Extracelulares , Degeneración del Disco Intervertebral , MicroARNs , Núcleo Pulposo , Plasma Rico en Plaquetas , Receptores CXCR , Animales , Apoptosis/genética , Vesículas Extracelulares/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/terapia , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Núcleo Pulposo/metabolismo , Plasma Rico en Plaquetas/metabolismo , Receptores CXCR/metabolismo , Factores de Transcripción SOXC/metabolismo
18.
Front Endocrinol (Lausanne) ; 13: 906586, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846294

RESUMEN

Chemokines, and their receptors play a crucial role in the pathophysiology of cardiovascular diseases (CVD). Chemokines classically mediate their effects by binding to G-protein-coupled receptors. The discovery that chemokines can also bind to atypical chemokine receptors (ACKRs) and initiate alternative signaling pathways has changed the paradigm regarding chemokine-related functions. Among these ACKRs, several studies have highlighted the exclusive role of ACKR3, previously known as C-X-C chemokine receptor type 7 (CXCR7), in CVD. Indeed, ACKR3 exert atheroprotective, cardioprotective and anti-thrombotic effects through a wide range of cells including endothelial cells, platelets, inflammatory cells, fibroblasts, vascular smooth muscle cells and cardiomyocytes. ACKR3 functions as a scavenger receptor notably for the pleiotropic chemokine CXCL12, but also as a activator of different pathways such as ß-arrestin-mediated signaling or modulator of CXCR4 signaling through the formation of ACKR3-CXCR4 heterodimers. Hence, a better understanding of the precise roles of ACKR3 may pave the way towards the development of novel and improved therapeutic strategies for CVD. Here, we summarize the structural determinant characteristic of ACKR3, the molecules targeting this receptor and signaling pathways modulated by ACKR3. Finally, we present and discuss recent findings regarding the role of ACKR3 in CVD.


Asunto(s)
Enfermedades Cardiovasculares , Receptores CXCR/metabolismo , Enfermedades Cardiovasculares/genética , Quimiocina CXCL12 , Células Endoteliales/metabolismo , Humanos , Transducción de Señal/fisiología
19.
Basic Res Cardiol ; 117(1): 30, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35674847

RESUMEN

Atherosclerosis is the foundation of potentially fatal cardiovascular diseases and it is characterized by plaque formation in large arteries. Current treatments aimed at reducing atherosclerotic risk factors still allow room for a large residual risk; therefore, novel therapeutic candidates targeting inflammation are needed. The endothelium is the starting point of vascular inflammation underlying atherosclerosis and we could previously demonstrate that the chemokine axis CXCL12-CXCR4 plays an important role in disease development. However, the role of ACKR3, the alternative and higher affinity receptor for CXCL12 remained to be elucidated. We studied the role of arterial ACKR3 in atherosclerosis using western diet-fed Apoe-/- mice lacking Ackr3 in arterial endothelial as well as smooth muscle cells. We show for the first time that arterial endothelial deficiency of ACKR3 attenuates atherosclerosis as a result of diminished arterial adhesion as well as invasion of immune cells. ACKR3 silencing in inflamed human coronary artery endothelial cells decreased adhesion molecule expression, establishing an initial human validation of ACKR3's role in endothelial adhesion. Concomitantly, ACKR3 silencing downregulated key mediators in the MAPK pathway, such as ERK1/2, as well as the phosphorylation of the NF-kB p65 subunit. Endothelial cells in atherosclerotic lesions also revealed decreased phospho-NF-kB p65 expression in ACKR3-deficient mice. Lack of smooth muscle cell-specific as well as hematopoietic ACKR3 did not impact atherosclerosis in mice. Collectively, our findings indicate that arterial endothelial ACKR3 fuels atherosclerosis by mediating endothelium-immune cell adhesion, most likely through inflammatory MAPK and NF-kB pathways.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Receptores CXCR , Animales , Aterosclerosis/metabolismo , Adhesión Celular , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Inflamación/metabolismo , Ratones , Ratones Noqueados para ApoE , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Receptores CXCR/metabolismo , Factor de Transcripción ReIA/metabolismo
20.
Int J Mol Sci ; 23(12)2022 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-35742971

RESUMEN

The accumulation of senescent cells in aging tissues is associated with age-related diseases and functional decline. Thus, senolysis, a therapy aimed at rejuvenation by removing senescent cells from the body, is being developed. However, this therapy requires the identification of membrane surface antigens that are specifically expressed on senescent cells for their selective elimination. We showed that atypical chemokine receptor 3 (ACKR3), a receptor of the CXC motif chemokine 12 (CXCL12) implicated in cancer, inflammation, and cardiovascular disorders, is selectively expressed on the surface of senescent human fibroblasts but not on proliferating cells. Importantly, the differential presence of ACKR3 enabled the isolation of senescent cells by flow cytometry using anti-ACKR3 antibodies. Furthermore, antibody-dependent cellular cytotoxicity assays revealed that cell surface ACKR3 preferentially sensitizes senescent but not dividing fibroblasts to cell injury by natural killer cells. Conclusively, the selective expression of ACKR3 on the surface of senescent cells allows the preferential elimination of senescent cells. These results might contribute to the future development of novel senolysis approaches.


Asunto(s)
Proteínas de la Membrana , Receptores CXCR/metabolismo , Senescencia Celular , Quimiocina CXCL12/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Células Asesinas Naturales/metabolismo , Proteínas de la Membrana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA