...
首页> 外文期刊>Cell death & disease. >Platelet-derived CXCL12 regulates monocyte function, survival, differentiation into macrophages and foam cells through differential involvement of CXCR4–CXCR7
【24h】

Platelet-derived CXCL12 regulates monocyte function, survival, differentiation into macrophages and foam cells through differential involvement of CXCR4–CXCR7

机译:血小板衍生的CXCL12通过CXCR4–CXCR7的不同参与来调节单核细胞功能,存活,分化为巨噬细胞和泡沫细胞

获取原文
   

获取外文期刊封面封底 >>

       

摘要

Platelets store and release CXCL12 (SDF-1), which governs differentiation of hematopoietic progenitors into either endothelial or macrophage-foam cells. CXCL12 ligates CXCR4 and CXCR7 and regulates monocyte/macrophage functions. This study deciphers the relative contribution of CXCR4–CXCR7 in mediating the effects of platelet-derived CXCL12 on monocyte function, survival, and differentiation. CXCL12 and macrophage migration inhibitory factor (MIF) that ligate CXCR4–CXCR7 induced a dynamic bidirectional trafficking of the receptors, causing CXCR4 internalization and CXCR7 externalization during chemotaxis, thereby influencing relative receptor availability, unlike MCP-1. In vivo we found enhanced accumulation of platelets and platelet-macrophage co-aggregates in peritoneal fluid following induction of peritonitis in mice. The relative surface expression of CXCL12, CXCR4, and CXCR7 among infiltrated monocytes was also enhanced as compared with peripheral blood. Platelet-derived CXCL12 from collagen-adherent platelets and recombinant CXCL12 induced monocyte chemotaxis specifically through CXCR4 engagement. Adhesion of monocytes to immobilized CXCL12 and CXCL12-enriched activated platelet surface under static and dynamic arterial flow conditions were mediated primarily through CXCR7 and were counter-regulated by neutralizing platelet-derived CXCL12. Monocytes and culture-derived-M1–M2 macrophages phagocytosed platelets, with the phagocytic potential of culture-derived-M1 macrophages higher than M2 involving CXCR4–CXCR7 participation. CXCR7 was the primary receptor in promoting monocyte survival as exerted by platelet-derived CXCL12 against BH3-mimetic induced apoptosis (phosphatidylserine exposure, caspase-3 activation, loss of mitochondrial transmembrane potential). In co-culture experiments with platelets, monocytes predominantly differentiated into CD163+ macrophages, which was attenuated upon CXCL12 neutralization and CXCR4/CXCR7 blocking antibodies. Moreover, OxLDL uptake by platelets induced platelet apoptosis, like other platelet agonists TRAP and collagen-related peptide (CRP). CXCL12 facilitated phagocytosis of apoptotic platelets by monocytes and M1–M2 macrophages, also promoted their differentiation into foam cells via CXCR4 and CXCR7. Thus, platelet-derived CXCL12 could regulate monocyte-macrophage functions through differential engagement of CXCR4 and CXCR7, indicating an important role in inflammation at site of platelet accumulation.
机译:血小板储存并释放CXCL12(SDF-1),后者控制造血祖细胞向内皮细胞或巨噬细胞泡沫细胞的分化。 CXCL12连接CXCR4和CXCR7,并调节单核细胞/巨噬细胞功能。这项研究破译了CXCR4–CXCR7在介导血小板衍生的CXCL12对单核细胞功能,存活和分化的影响中的相对作用。结扎CXCR4–CXCR7的CXCL12和巨噬细胞迁移抑制因子(MIF)诱导了受体的动态双向转运,从而在趋化过程中引起CXCR4内在化和CXCR7外在化,从而影响相对受体的可用性,与MCP-1不同。在体内,我们发现诱导小鼠腹膜炎后,血小板和血小板-巨噬细胞共聚集体在腹膜液中的积累增加。与外周血相比,浸润的单核细胞之间CXCL12,CXCR4和CXCR7的相对表面表达也有所增强。来自胶原蛋白粘附的血小板的血小板衍生的CXCL12和重组CXCL12特别通过CXCR4参与诱导单核细胞趋化性。在静态和动态动脉血流条件下,单核细胞对固定化CXCL12和富含CXCL12的活化血小板表面的粘附主要通过CXCR7介导,并通过中和血小板衍生的CXCL12进行反调节。单核细胞和培养物衍生的M1–M2巨噬细胞吞噬血小板,而培养物衍生的M1巨噬细胞的吞噬潜能高于参与CXCR4–CXCR7参与的M2。 CXCR7是促进单核细胞存活的主要受体,如血小板衍生的CXCL12对抗BH3模拟诱导的凋亡(磷脂酰丝氨酸暴露,caspase-3活化,线粒体跨膜电位丧失)。在与血小板的共培养实验中,单核细胞主要分化为CD163 + 巨噬细胞,其在CXCL12中和和CXCR4 / CXCR7阻断抗体后会减弱。此外,与其他血小板激动剂TRAP和胶原相关肽(CRP)一样,血小板摄取OxLDL也会诱导血小板凋亡。 CXCL12促进单核细胞和M1-M2巨噬细胞对凋亡性血小板的吞噬作用,也促进它们通过CXCR4和CXCR7分化为泡沫细胞。因此,血小板衍生的CXCL12可以通过CXCR4和CXCR7的不同结合来调节单核巨噬细胞功能,这在血小板积聚部位的炎症中起重要作用。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号