首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 125 毫秒
1.
为探讨CXC趋化因子8(CXCL8)及其受体CXC趋化因子受体1(CXCR1)、CXC趋化因子受体2(CXCR2)在慢性乙型肝炎中的表达及意义,本研究选取了我院治疗的慢性乙型肝炎患者64例(观察组),同时选取健康志愿者60例作为对照组,两组均采用SABC免疫细胞化学染色法检测CXCL8、CXCR1和CXCR2在各组外周血中性粒细胞(PMNs)内的表达量,采用RT-PCR检测PMNs中CXCL8、CXCR1和CXCR2mRNA表达。实验发现,观察组外周血PMNs中CXCL8、CXCR1表达明显强于对照组(p0.05);观察组和对照组外周血PMNs中CXCR2表达强度差异比较无统计学意义(p0.05);观察组外周血PMNs中CXCL8mRNA、CXCR1 mRNA和CXCR2 mRNA相对表达量分别为(1.16±0.15)、(0.87±0.24)和(1.01±0.22),明显高于对照组(p0.05);观察组中HBV-DNA阳性者外周血PMNs中CXCL8 mRNA和CXCR1 mRNA相对表达量分别为(1.27±0.10)和(1.02±0.13),明显高于HBV-DNA阴性者(p0.05);HBV-DNA阳性者和HBV-DNA阴性者CXCR2 mRNA相对表达量比较差异无统计学意义(p0.05);CXCL8 mRNA和CXCR1 mRNA相对表达量与ALT呈正相关(r=0.673和0.681,p0.05),CXCR2 mRNA相对表达量与ALT无相关性(p0.05)。慢性乙肝患者PMNs内CXCL8、CXCR1、CXCR2的mRNA水平升高,其中CXCL8、CXCR1的mRNA与血清ALT呈正相关,同时与HBV DNA载量有一定的相关性。  相似文献   

2.
目的:探讨MDA-MB-231细胞经无血清培养富集三阴性乳腺癌干细胞,观察再成球、集落形成及CD44+CD24-/low、CXCR4表达。方法:将MDA-MB-231乳腺癌细胞进行微球体培养,取培养第7-9天的微球体,判断干细胞富集的程度;比较不同细胞浓度对癌球细胞成球率影响;流式细胞仪测定CD44+CD24-/low含量;Western blot法分析CXCR4蛋白表达;单个癌球细胞再成球能力;观察癌球与贴壁细胞集落形成。结果:1).在含20 ng/m L EGF,10 ng/m L b FGF,2%b27无血清培养基中可培养三阴性乳腺癌癌球,1×104/m L、2×104/m L、3×104/m L、4×104/m L、5×104/m L细胞浓度癌球细胞成球率分别为(5.61±0.02)%、(3.23±0.54)%、(2.28±0.48)%、(1.05±0.13)%、(0.91±0.01)%,组间比较差异有统计学意义P值均0.05。2).贴壁MDA-MB-231细胞与癌球细胞CD44+CD24-/low含量分别为(38.54±2.00)%VS(66.35±2.06)%,差异有统计学意义P=0.003。3).癌球细胞CXCR4蛋白表达高于贴壁MDA-MB-231细胞,灰度扫描分析差异有统计学意义,P=0.03。4).单个癌球细胞具有再成球能力。5).软琼脂糖集落形成能力癌球需200个细胞即可见集落形成,而贴壁细胞需1 000个MDA-MB-231细胞。结论:1.通过无血清培养可以富集三阴性乳腺癌干细胞,低细胞密度有利于癌球形成。2.癌球中CD44+CD24-/low含量高于贴壁MDA-MB-231细胞。3.CXCR4在癌球中表达高于贴壁MDA-MB-231细胞。  相似文献   

3.
目的:检测系统性红斑狼疮(systemic lupus erythematosus,SLE)合并带状疱疹患者外周血CD4~+CD28~+和CD4~+CD25~+Fox P3~+调节性T细胞的表达及相关性,探讨其在SLE合并带状疱疹发病中的临床意义。方法:采用流式细胞术检测30例SLE患者、30例SLE合并带状疱疹患者及30例健康对照者外周血中CD4~+/CD8~+T淋巴细胞亚群表面CD28的表达及CD4~+CD25~+Fox P3~+Treg细胞的表达水平,并分析SLE合并带状疱疹患者外周血CD4~+CD28~+和CD4~+CD25~+Fox P3~+调节性T细胞表达的相关性。结果:SLE合并带状疱疹组患者急性期外周血CD4~+T淋巴细胞比率、绝对计数显著降低,CD4~+、CD8~+T淋巴细胞表面的CD28表达下调,CD4~+CD25~+Fox P3~+Treg细胞水平显著高于SLE组及健康对照组,SLE合并带状疱疹组患者外周血CD4~+CD25~+Fox P3~+Treg水平与CD4~+CD28~+水平成负相关(P均0.05)。结论:SLE合并带状疱疹患者CD4~+、CD8~+T细胞活化异常,CD4~+CD25~+Fox P3~+Treg细胞可能参与抑制了T细胞的活化。  相似文献   

4.
目的:探讨HIV/HCV重叠感染患者外周血单核细胞亚群与肝损伤的关系。方法:观察对象为HIV/HCV重叠感染患者,分为对照组(n=11)、肝纤维化组(n=12)和肝硬化组(n=7)。运用流式细胞仪检测单核细胞及其亚群变化,瞬时弹性成像(Fibroscan)检测肝纤维化情况。比较单核细胞各亚群在不同程度肝损伤中的差异,并对HIV/HCV重叠感染患者外周血的单核细胞数与肝纤维化情况进行相关性分析。结果:HIV/HCV重叠感染患者肝硬化组与对照组比较,单核细胞CD14low CD16+和CD14high CD16+亚群显著增多(P=0.047,P=0.018)。HIV/HCV重叠感染患者肝纤维化组与对照组比较,单核细胞各亚群差异无统计学意义(P=0.84,P=0.812)。HIV/HCV重叠感染患者CD14high CD16+单核细胞与肝纤维化情况存在正性线性相关,方程成立,并且系数有统计学意义(P=0.018),方程似然比(r 2)0.45。结论:HIV/HCV重叠感染患者CD14high CD16+单核细胞增高有可能是肝损伤加重的原因之一。  相似文献   

5.
目的:探讨CXCR4与MMP-9在膀胱移行细胞癌中表达的相关性及其临床意义.方法:采用免疫组化SP法与半定量RT-PCR检测40例膀胱移行细胞癌组织及10例正常膀胱粘膜组织中CXCR4和MMP-9蛋白及mRNA的表达情况,分析膀胱移行细胞癌组织中CXCR4和MMP-9表达的相关性,分析二者与临床病理特征的关系.结果:膀胱移行细胞癌组织中CXCR4蛋白表达率为77.5%,mRNA相对含量为0,777±0.044;其中浸润深度达肌层者表达率为100%,mRNA相对含量为0.790± 0.049;局限在粘膜下层者表达率为50%,mRNA相对含量为0.660± 0.052;二者之间差异有统计学意义.MMP-9在膀胱移行细胞癌组织中的表达率为80.0%,mRNA相对含量为0.850± 0.079,其中浸润深度达肌层者表达率为95.5%,mRNA相对含量为0.854±0.070,局限在粘膜下层者表达率为61.1%,mRNA相对含量为0.758±0.092,二者之间差异有统计学意义.膀胱移行细胞癌组织中CXCR4及MMP-9蛋白阳性表达呈正相关关系(γ=0.479,P<0.05).MMP-9与肿瘤组织学分级有关,与患者的性别、年龄无关;而CXCR4的表达与肿瘤组织学分级及患者的性别、年龄均无关.结论:CXCR4和MMP-9表达与膀胱移行细胞癌的发生和浸润密切相关,通过干预CXCR4和MMP-9的活性可能成为治疗膀胱移行细胞癌的新靶点.  相似文献   

6.
目的:研究滤泡辅助性T细胞(T follicular helper cells,Tfh)在免疫性血小板减少性紫癜(immune thrombocytopenic purpura,ITP)患者的表达并探讨其临床意义。方法:用流式细胞术检测20例健康人、25例ITP患者外周血CXCR5+CD4+T细胞占CD4+T细胞的比例。结果:与健康对照组相比,ITP患者外周血CXCR5+CD4+T细胞占CD4+T细胞的比例显著增高(P<0.05)。结论:Tfh在ITP患者外周血比例增高,为Tfh能否为ITP的免疫调节和干预提出新的方向提供了证据。  相似文献   

7.
研究趋化因子受体4(Chemokine receptor 4,CXCR4)的表达水平与骨肉瘤肺转移的关系并探讨CXCR4在骨肉瘤组织中表达的意义.采用免疫组织化学方法测定5例肺转移和11例无肺转移患者的骨肉瘤组织中CXCR4的表达水平.发现16例骨肉瘤组织中均有CXCR4蛋白的表达,但其表达水平存在差异,与肺转移的骨肉瘤组织(80.52±9.93)相比,11例非肺转移的骨肉瘤组织中CXCR4的表达水平(65.56±12.75)显著较低(P=0.037<0.05).实验结果表明CXCR4的上调表达见于肺转移率较高的骨肉瘤组织,提示CXCR4可能参与了骨肉瘤的肺转移.  相似文献   

8.
目的:探讨SDF-1/CXCR4及VEGF-C在喉癌淋巴结转移中的作用机制。方法:随机选取2012年8月至2015年8月我院收治的90例喉癌患者,将这些患者作为研究组,另选取20例具有相应正常粘膜组织的患者为对照组,运用免疫组化SP法对CXCR4及VEGF-C及SDF-1进行检测,分析SDF-1/CXCR4及VEGF-C在喉癌淋巴结组织中的表达及其与临床病理特征的关系。结果:喉癌组织中CXCR4、VEGF-C、SDF-1的阳性表达率均显著高于正常组织(P0.05);Ⅲ+Ⅳ患者CXCR4、VEGF-C、SDF-1的阳性表达率均显著高于Ⅰ+Ⅱ患者(P0.05);低分化患者CXCR4、VEGF-C的阳性表达率均显著高于高中分化患者(P0.05),但SDF-1阳性表达率之间比较,差异均不具有统计学意义(P0.05);淋巴结转移患者CXCR4、VEGF-C、SDF-1的阳性表达率均显著高于未发生淋巴结转移的患者,差异具有统计学意义(P0.05);不同年龄、病变部位患者CXCR4、VEGF-C、SDF-1阳性表达率之间比较,差异均不具有统计学意义(P0.05);喉癌组织中CXCR4及VEGF-C阳性表达均呈显著的正相关关系(P0.05);阴性表达也均呈显著的正相关关系(P0.05)。结论:SDF-1/CXCR4及VEGF-C在喉癌淋巴结转移中高表达,可能共同促进喉癌淋巴结转移。  相似文献   

9.
C-X-C趋化因子受体4(CXCR4)是乳腺癌细胞运动的关键调节因子。CXCR4的功能性表达与乳腺癌的恶性进展密切相关。酪氨酸硫酸化转移酶1(tyrosylprotein sulfotransferase 1,TPST1)是CXCR4蛋白翻译后酪氨酸硫酸化修饰的一个关键酶。本研究将探索TPST1在CXCR4调节乳腺癌细胞侵袭过程中的作用机制。利用定量PCR,免疫组织化学和蛋白质免疫印迹等试验技术检测乳腺癌组织和细胞系中CXCR4和TPST1的mRNA和蛋白表达水平。RNA干扰,趋化试验和侵袭试验用于检测TPST1对于CXCR4诱导的乳腺癌细胞侵袭的影响。研究发现CXCR4蛋白在乳腺癌转移淋巴结组织中呈高表达(P=0. 0016)。CXCR4在乳腺癌转移淋巴结组织中的高表达与肿瘤浸润深度密切相关(P=0. 026)。TPST1与CXCR4蛋白表达在乳腺癌原发组织和配对转移淋巴结组织中均呈显著正相关(P=0. 009; P=0. 006)。TPST1在高度恶性乳腺癌MDA-MB-231细胞中呈高表达,在低度恶性乳腺癌MCF-7细胞中弱表达,而两者CXCR4表达基本相同。小RNA干扰降低TPST1的表达后,下调了乳腺癌MDA-MB-231细胞对于CXCR4配体即基质细胞衍生因子1α(stromal cell-derived factor 1 alpha,SDF-1α)的运动反应性,进而降低CXCR4诱导的MDA-MB-231细胞迁移和侵袭能力。综上,在CXCR4诱导的乳腺癌细胞侵袭过程中,TPST1表达对于CXCR4功能性活化至关重要,TPST1可能作为潜在的抗CXCR4药物治疗乳腺癌恶性进展的联合靶点。  相似文献   

10.
目的:探讨c AMP反应原件结合蛋白(cyclic AMP response element-binding protein,CREB)和CXC趋化因子受体2(CXC chemokine receptor 2,CXCR2)在非小细胞肺癌(non-small cell lung cancer,NSCLC)组织中的表达及与肺癌发生、发展的关系。方法:采用免疫组化Elivision法检测CREB和CXCR2在49例NSCLC组织中的表达,同时随机选取20例癌旁正常肺组织作为对照。结果:NSCLC中CREB和CXCR2的表达阳性率分别为57.1%和69.4%,均显著高于癌旁正常组织中的10.0%和15.0%(P0.05);同时,CREB和CXCR2在NSCLC组织中的表达不仅与肿瘤的TNM分期有关(P0.05),而且还与肿瘤的分化程度有关(P0.05);另外,CREB在吸烟患者中的表达明显高于非吸烟患者(P0.05),在鳞癌组织中的表达明显高于腺癌(P0.05);CREB和CXCR2的表达呈正相关。结论:CREB和CXCR2可能参与了NSCLC的发生和发展,为NSCLC的靶向治疗提供了新的依据。  相似文献   

11.
Although chemokines are well established to function in immunity and endothelial cell activation and proliferation, a rapidly growing literature suggests that CXC Chemokine receptors CXCR3, CXCR4 and CXCR7 are critical in the development and progression of solid tumors. The effect of these chemokine receptors in tumorigenesis is mediated via interactions with shared ligands I-TAC (CXCL11) and SDF-1 (CXCL12). Over the last decade, CXCR4 has been extensively reported to be overexpressed in most human solid tumors and has earned considerable attention toward elucidating its role in cancer metastasis. To enrich the existing armamentarium of anti-cancerous agents, many inhibitors of CXCL12–CXCR4 axis have emerged as additional or alternative agents for neo-adjuvant treatments and even many of them are in preclinical and clinical stages of their development. However, the discovery of CXCR7 as another receptor for CXCL12 with rather high binding affinity and recent reports about its involvement in cancer progression, has questioned the potential of “selective blockade” of CXCR4 as cancer chemotherapeutics. Interestingly, CXCR7 can also bind another chemokine CXCL11, which is an established ligand for CXCR3. Recent reports have documented that CXCR3 and their ligands are overexpressed in different solid tumors and regulate tumor growth and metastasis. Therefore, it is important to consider the interactions and crosstalk between these three chemokine receptors and their ligand mediated signaling cascades for the development of effective anti-cancer therapies. Emerging evidence also indicates that these receptors are differentially expressed in tumor endothelial cells as well as in cancer stem cells, suggesting their direct role in regulating tumor angiogenesis and metastasis. In this review, we will focus on the signals mediated by this receptor trio via their shared ligands and their role in tumor growth and progression.  相似文献   

12.
中性粒细胞属非特异性免疫细胞,其表面可表达CXCR1和CXCR2.IL-8是其共同配体,它们彼此结合激活后续级联信号传导,产生一系列生物学效应,在介导炎症反应、促进血管新生、维持中性粒细胞稳态等起重要作用.Reparixin是非竞争变构的CXCR1和CXCR2阻滞剂,可抑制中性粒细胞过度趋化、迁移介导的炎症反应.  相似文献   

13.
CXCR4 and CXCR7 have distinct functions in regulating interneuron migration   总被引:1,自引:0,他引:1  
CXCL12/CXCR4 signaling is critical for cortical interneuron migration and their final laminar distribution. No information is yet available on CXCR7, a newly defined CXCL12 receptor. Here we demonstrated that CXCR7 regulated interneuron migration autonomously, as well as nonautonomously through its expression in immature projection neurons. Migrating cortical interneurons coexpressed Cxcr4 and Cxcr7, and Cxcr7(-/-) and Cxcr4(-/-) mutants had similar defects in interneuron positioning. Ectopic CXCL12 expression and pharmacological blockade of CXCR4 in Cxcr7(-/-) mutants showed that both receptors were essential for responding to CXCL12 during interneuron migration. Furthermore, live imaging revealed that Cxcr4(-/-) and Cxcr7(-/-) mutants had opposite defects in interneuron motility and leading process morphology. In?vivo inhibition of Gα(i/o) signaling in migrating interneurons phenocopied the interneuron lamination defects of Cxcr4(-/-) mutants. On the other hand, CXCL12 stimulation of CXCR7, but not CXCR4, promoted MAP kinase signaling. Thus, we suggest that CXCR4 and CXCR7 have distinct roles and signal transduction in regulating interneuron movement and laminar positioning.  相似文献   

14.
袁泉  许丞  张翔  卢东  张捷 《现代生物医学进展》2016,16(27):5273-5275
目的:探讨膀胱癌组织中趋化因子受体4(CXCR4)和趋化因子受体7(CXCR7)的表达及临床意义。方法:收集2012年1月至2014年1月我院收集的膀胱癌组织标本96例,肿瘤旁正常组织标本42例,采用免疫组化方法检测组织标本中CXCR4和CXCR7的表达情况。结果:96例癌组织中检出CXCR4阳性59例,阳性率为61.46%,检出CXCR7阳性表达71例,阳性率为73.96%;42例癌旁组织中检出CXCR4阳性11例,阳性率26.19%,检出CXCR7阳性8例,阳性率为19.05%,癌组织与癌旁组织中CXCR4和CXCR7的表达具有统计学差异(均P0.05);相关性分析显示在膀胱癌组织中,CXCR4和CXCR7的表达呈正相关性(r=0.497,P=0.001);CXCR4和CXCR7在浸润性高(T2-T3)的膀胱癌和分化程度低(G2-G3)的膀胱癌表达强度较高,且差异具有统计学意义(均P0.05)。结论:CXCR4和CXCR7协同参与了膀胱癌的发生发展,并且与肿瘤的分化程度和浸润程度密切相关,有望成为诊断和治疗的重要靶点,在临床应用上具有重要意义。  相似文献   

15.
Treatment of metastatic renal cell carcinoma (mRCC) has improved significantly with the advent of agents targeting the mTOR pathway, such as temsirolimus and everolimus. However, their efficacy is thought to be limited by feedback loops and crosstalk with other pathways leading to the development of drug resistance. As CXCR4–CXCL12–CXCR7 axis has been described to have a crucial role in renal cancer; the crosstalk between the mTOR pathway and the CXCR4–CXCL12–CXCR7 chemokine receptor axis has been investigated in human renal cancer cells. In SN12C and A498, the common CXCR4–CXCR7 ligand, CXCL12, and the exclusive CXCR7 ligand, CXCL11, activated mTOR through P70S6K and 4EBP1 targets. The mTOR activation was specifically inhibited by CXCR4 antagonists (AMD3100, anti-CXCR4-12G5 and Peptide R, a newly developed CXCR4 antagonist) and CXCR7 antagonists (anti-CXCR7-12G8 and CCX771, CXCR7 inhibitor). To investigate the functional role of CXCR4, CXCR7 and mTOR in human renal cancer cells, both migration and wound healing were evaluated. SN12C and A498 cells migrated toward CXCL12 and CXCL11; CXCR4 and CXCR7 inhibitors impaired migration and treatment with mTOR inhibitor, RAD001, further inhibited it. Moreover, CXCL12 and CXCL11 induced wound healing while was impaired by AMD3100, the anti CXCR7 and RAD001. In SN12C and A498 cells, CXCL12 and CXCL11 promoted actin reorganization characterized by thin spikes at the cell periphery, whereas AMD3100 and anti-CXCR7 impaired CXCL12/CXCL11-induced actin polymerization, and RAD001 treatment further reduced it. In addition, when cell growth was evaluated in the presence of CXCL12, CXCL11 and mTOR inhibitors, an additive effect was demonstrated with the CXCR4, CXCR7 antagonists and RAD001. RAD001-resistant SN12C and A498 cells recovered RAD001 sensitivity in the presence of CXCR4 and CXCR7 antagonists. In conclusion, the entire axis CXCR4–CXCL12–CXCR7 regulates mTOR signaling in renal cancer cells offering new therapeutic opportunities and targets to overcome resistance to mTOR inhibitors.Renal cell carcinoma (RCC) is the most lethal malignancy among urological cancers with a total of 64 770 new cases and 13 570 deaths estimated in the United States in 2012.1 A growing understanding of the molecular biology of RCC changed the therapeutic approach toward target-based agents. Since 2005, the US Food and Drug Administration (FDA) has approved six new target agents for metastatic RCC that antagonize two principal signaling pathways: the vascular endothelial growth factor receptor (VEGF) and the mammalian target of rapamycin (mTOR).2 The mTOR is an atypical intracellular serine/threonine protein kinase regulated by phosphatidylinositol 3-kinase (PI3K).3 mTOR exists in two distinct complexes termed mTOR complex 1 (mTORC1) comprising mTOR, mLST8 (also termed G-protein β-subunit-like protein, GβL, a yeast homolog of LST8), raptor (regulatory associated protein of mTOR) and PRAS40 (proline-rich Akt substrate, 40 kDa), and mTOR complex 2 (mTORC2) comprising mTOR, mLST8, rictor (rapamycin-insensitive companion of mTOR), mSin1 (mammalian stress-activated protein kinase (SAPK)-interacting protein 1), protor (protein observed with rictor) and PRR5 (proline-rich protein 5).4 mTORC1 responds to amino acids, stress, oxygen, energy and growth factors and is sensitive to rapamycin; when active, mTORC1 promotes cell growth and also drives cell-cycle progression. Alternatively, mTORC2 regulates cytoskeletal organization and cell survival/metabolism and is sensitive to rapamycin over longer incubation times or at higher doses.3 mTORC1 controls cell growth and translation through the phosphorylation of ribosomal protein S6 kinase (S6K) and of eukaryotic translation initiation factor 4EBP1, which regulate either the translation of ribosomal proteins or the cap-dependent translation by inhibition of eukaryotic translation initiation factor 4E, respectively.3, 4 The activated mTOR pathway has been identified in several human malignancies, thus being an attractive target for anticancer therapy. mTORC1 activity is inhibited by rapalogs such as rapamycin (sirolimus) and associated analogs (temsirolimus/CCI-779, RAD001, ridaforolimus/AP23573).5 These drugs suppress mTORC1 activity forming a complex with FK506-binding protein 12. Temsirolimus (rapamycin analog) was the first mTOR inhibitor approved as first-line treatment in patients with poor-prognosis metastatic RCC (mRCC) patients,3 ridaforolimus is currently tested in phase III clinical trials5 and RAD001 is indicated as second-line treatment in patients with RCC at failure of first-line treatment with sunitinib or sorafenib. Other indications are subependymal giant cell astrocytoma associated with tuberous sclerosis and progressive neuroendocrine tumors of pancreatic origin.5 Although mTOR inhibitors prolong progression-free survival in patients with advanced RCC, most patients develop resistance to mTOR-inhibiting agents, limiting their efficacy; the new frontier of inhibiting the mTOR pathway is to identify agents targeting the feedback loops and crosstalks with other pathways involved in the acquired resistance to mTOR inhibitors.6Chemokines and their receptors have been implicated in regulating RCC growth, angiogenesis and metastases.7 In RCC, VHL mutation resulted in HIF-dependent CXCR4 activation8 and CXCR4 expression predicted poor tumor-specific survival.8, 9, 10 Recently, CXCL12 was shown to bind with high affinity the orphan receptor CXCR7/RDC1, which also binds a second ligand in the form of interferon-inducible T-cell α chemoattractant (I-TAC/CXCL11).11 Whereas the CXCR4 activity is primarily G-protein-mediated, CXCR7 is considered an atypical GPCR because ligand binding does not result in intracellular Ca2+ release.11 Some studies provided evidence that CXCR7 represents a ‘decoy'' receptor, which is responsible for either sequestering extracellular CXCL1212 or modulating CXCR4 signaling by forming CXCR7–CXCR4 heterodimers.13 In contrast, others demonstrated that CXCR7 relays intracellular signals14, 15, 16, 17 and promotes cell motility18, 13, 19 acting through β-arrestin.20, 21 CXCR7 is highly expressed in human cancers such as prostate, lung, glioma, ovarian, breast cancer cells and in tumor-associated blood vessels and seems to be essential for survival, adhesion and growth of tumor cells.11, 14, 15, 22, 23, 24 It was recently demonstrated that CXCR4 and CXCR7 predict prognosis in RCC.10, 25 CXCL12 activates CXCR4 and the derived signaling can transduce on the mTOR pathway in pancreatic cancer, gastric cancer and T-cell leukemia cells;26, 27, 28, 29 antagonists targeting PI3K and/or mTOR inhibited CXCL12-mediated cell migration and this effect was primarily attributed to the inhibition of mTORC1 and consequent decrease in RhoA, Cdc42 and Rac1 in human gastric carcinoma cells.28Aim of the study was to evaluate interactions between the CXCL12–CXCR4–CXCR7 axis and the mTOR pathway in human renal cancer cells to identify new therapeutic opportunities and overcome resistance mechanisms.  相似文献   

16.
Gallbladder carcinoma (GBC) is a vicious and invasive disease. The major challenge in the clinical treatment of GBC is the lack of a suitable prognosis method. Chemokine receptors such as CXCR3, CXCR4 and CXCR7 play vital roles in the process of tumour progression and metastasis. Their expression levels and distribution are proven to be indicative of the progression of GBC, but are hard to be decoded by conventional pathological methods, and therefore, not commonly used in the prognosis of GBC. In this study, we developed a computer‐aided image analysis method, which we used to quantitatively measure the expression levels of CXCR3, CXCR4 and CXCR7 in the nuclei and cytoplasm of glandular and interstitial cells from a cohort of 55 GBC patients. We found that CXCR3, CXCR4 and CXCR7 expressions are associated with the clinicopathological variables of GBC. Cytoplasmic CXCR3, nuclear CXCR7 and cytoplasmic CXCR7 were significant predictive factors of histology invasion, whereas cytoplasmic CXCR4 and nuclear CXCR4 were significantly correlated with T and N stage and were associated with the overall survival and disease‐free survival. These results suggest that the quantification and localisation of CXCR3, CXCR4 and CXCR7 expressions in different cell types should be considered using computer‐aided assessment to improve the accuracy of prognosis in GBC.  相似文献   

17.
Chemokines are secreted into the tumor microenvironment by tumor-infiltrating inflammatory cells as well as by tumor cells. Chemokine receptors mediate agonist-dependent cell responses, including migration and activation of several signaling pathways. In the present study we show that several human melanoma cell lines and melanoma cells on macroscopically infiltrated lymph nodes express the chemokine receptors CXCR3 and CXCR4. Using the highly invasive melanoma cell line BLM, we demonstrate that the chemokine Mig, a ligand for CXCR3, activates the small GTPases RhoA and Rac1, induces a reorganization of the actin cytoskeleton, and triggers cell chemotaxis and modulation of integrin VLA-5- and VLA-4-dependent cell adhesion to fibronectin. Furthermore, the chemokine SDF-1alpha, the ligand of CXCR4, triggered modulation of beta(1) integrin-dependent melanoma cell adhesion to fibronectin. Additionally, Mig and SDF-1alpha activated MAPKs p44/42 and p38 on melanoma cells. Expression of functional CXCR3 and CXCR4 receptors on melanoma cells indicates that they might contribute to cell motility during invasion as well as to regulation of cell proliferation and survival.  相似文献   

18.
19.
A series of N-(2-hydroxy-3-sulfonamidobenzene)-N'-arylcyanoguanidines was prepared. In general, these compounds proved to be potent antagonists of CXCR2 while the selectivity versus CXCR1 ranged from non-selective to >200-fold.  相似文献   

20.
Stimulation of microvascular endothelial cells with interleukin (IL)-8 leads to cytoskeletal reorganization, which is mediated by combined activation of the CXCR1 and the CXCR2. In the early phase actin stress fibers appear, followed by cortical actin accumulation and cell retraction leading to gap formation between cells. The early response (between 1 and 5 min) is inhibited by an antibody that blocks the CXCR1. The later phase (from about 5 to 60 min), which is associated with cell retraction, is prevented by anti-CXCR2 antibody. Furthermore, anti-CXCR2, but not anti-CXCR1, antibody blocked IL-8-mediated haptotaxis of endothelial cells on collagen. The later phase of the IL-8-mediated actin response is inhibited by pertussis toxin, indicating that the CXCR2 couples to G(i). In contrast, the early phase is blocked by C3 botulinum toxin, which inactivates Rho, and by Y-27632, which inhibits Rho kinase, but not by pertussis toxin. Furthermore, the early CXCR1-mediated formation of stress fibers was prevented by dominant negative Rho. Dominant negative Rac on the other hand initially translocated to actin-rich filopodia after stimulation with IL-8 and later prevented cell retraction by blocking the CXCR2-mediated cytoskeletal response. These results indicate that IL-8 activates both the CXCR1 and the CXCR2 on microvascular endothelial cells, using different signal transduction cascades. The retraction of endothelial cells due to activation of the CXCR2 may contribute to the increased vascular permeability observed in acute inflammation and during the angiogenic response.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号