...
首页> 外文期刊>The Journal of biological chemistry >Transient Receptor Potential Mucolipin 1 (TRPML1) and Two-pore Channels Are Functionally Independent Organellar Ion Channels
【24h】

Transient Receptor Potential Mucolipin 1 (TRPML1) and Two-pore Channels Are Functionally Independent Organellar Ion Channels

机译:瞬态受体潜在的粘液1(TRPML1)和双孔通道是功能独立的细胞细胞离子通道

获取原文

摘要

NAADP is a potent second messenger that mobilizes Ca2+ from acidic organelles such as endosomes and lysosomes. The molecular basis for Ca2+ release by NAADP, however, is uncertain. TRP mucolipins (TRPMLs) and two-pore channels (TPCs) are Ca2+-permeable ion channels present within the endolysosomal system. Both have been proposed as targets for NAADP. In the present study, we probed possible physical and functional association of these ion channels. Exogenously expressed TRPML1 showed near complete colocalization with TPC2 and partial colocalization with TPC1. TRPML3 overlap with TPC2 was more modest. TRPML1 and to some extent TRPML3 co-immunoprecipitated with TPC2 but less so with TPC1. Current recording, however, showed that TPC1 and TPC2 did not affect the activity of wild-type TRPML1 or constitutively active TRPML1(V432P). N-terminally truncated TPC2 (TPC2delN), which is targeted to the plasma membrane, also failed to affect TRPML1 and TRPML1(V432P) channel function or TRPML1(V432P)-mediated Ca2+ influx. Whereas overexpression of TPCs enhanced NAADP-mediated Ca2+ signals, overexpression of TRPML1 did not, and the dominant negative TRPML1(D471K) was without affect on endogenous NAADP-mediated Ca2+ signals. Furthermore, the single channel properties of NAADP-activated TPC2delN were not affected by TRPML1. Finally, NAADP-evoked Ca2+ oscillations in pancreatic acinar cells were identical in wild-type and TRPML1?/? cells. We conclude that although TRPML1 and TPCs are present in the same complex, they function as two independent organellar ion channels and that TPCs, not TRPMLs, are the targets for NAADP.
机译:Naadp是一种有效的第二个信使,可使Ca2 +从酸性细胞器如臭氧和溶酶体中动员。然而,NaADP的CA2 +释放的分子基础是不确定的。 TRP粘蛋白(TRPMLS)和双孔通道(TPC)是存在于底糖系统内的CA2 +可-ETMETET离子通道。两者都被提出为Naadp的目标。在本研究中,我们探讨了这些离子通道的可能的物理和功能关系。外源表达的TRPML1显示出与TPC2的完全分层化和TPC1的部分分层化。 TRPML3与TPC2重叠更为适度。 TRPML1以及某种程度范围TRPML3与TPC2共沉淀,但TPC1较少。然而,目前的记录显示TPC1和TPC2不影响野生型TRPML1或组成型活性TRPML1(V432P)的活动。靶向血浆膜的N-末端截短的TPC2(TPC2DELN)也未能影响TRPML1和TRPML1(V432P)信道功能或TRPML1(V432P)介导的CA2 +流入。而TPCS增强的NaADP介导的CA2 +信号的过表达,TRPML1的过度表达没有,并且显性负TrpML1(D471K)没有影响内源Naadp介导的Ca2 +信号。此外,NaADP激活的TPC2Deln的单一信道属性不受TRPML1的影响。最后,在野生型和TRPML1中胰腺醋细胞中的Naadp诱发的Ca2 +振荡是相同的?/?细胞。我们得出结论,尽管TRPML1和TPC存在于同一复合物中,但它们用作两个独立的细胞内离子通道,并且TPC不是TRPMLS是NaADP的目标。

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