首页 | 本学科首页   官方微博 | 高级检索  
     检索      


In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15
Authors:Lisa Seipold  Hermann Altmeppen  Tomas Koudelka  Andreas Tholey  Petr Kasparek  Radislav Sedlacek  Michaela Schweizer  Julia Bär  Marina Mikhaylova  Markus Glatzel  Paul Saftig
Institution:1.Institute of Biochemistry,Christian Albrechts University Kiel,Kiel,Germany;2.Institute of Neuropathology,University Medical Center Hamburg-Eppendorf,Hamburg,Germany;3.Systematic Proteome Research and Bioanalytics, Institute for Experimental Medicine,Christian Albrechts University Kiel,Kiel,Germany;4.Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Division BIOCEV,Institute of Molecular Genetics of the CAS, v. v. i,Vestec,Czech Republic;5.Center for Molecular Neurobiology Hamburg (ZMNH),University Medical Center Hamburg-Eppendorf (UKE),Hamburg,Germany;6.Center for Molecular Neurobiology Hamburg (ZMNH), Emmy-Noether Group “Neuronal Protein Transport”,University Medical Center Hamburg-Eppendorf (UKE),Hamburg,Germany
Abstract:A disintegrin and metalloproteinase 10 (ADAM10) plays a major role in the ectodomain shedding of important surface molecules with physiological and pathological relevance including the amyloid precursor protein (APP), the cellular prion protein, and different cadherins. Despite its therapeutic potential, there is still a considerable lack of knowledge how this protease is regulated. We have previously identified tetraspanin15 (Tspan15) as a member of the TspanC8 family to specifically associate with ADAM10. Cell-based overexpression experiments revealed that this binding affected the maturation process and surface expression of the protease. Our current study shows that Tspan15 is abundantly expressed in mouse brain, where it specifically interacts with endogenous ADAM10. Tspan15 knockout mice did not reveal an overt phenotype but showed a pronounced decrease of the active and mature form of ADAM10, an effect which augmented with aging. The decreased expression of active ADAM10 correlated with an age-dependent reduced shedding of neuronal (N)-cadherin and the cellular prion protein. APP α-secretase cleavage and the expression of Notch-dependent genes were not affected by the loss of Tspan15, which is consistent with the hypothesis that different TspanC8s cause ADAM10 to preferentially cleave particular substrates. Analyzing spine morphology revealed no obvious differences between Tspan15 knockout and wild-type mice. However, Tspan15 expression was elevated in brains of an Alzheimer’s disease mouse model and of patients, suggesting that upregulation of Tspan15 expression reflects a cellular response in a disease state. In conclusion, our data show that Tspan15 and most likely also other members of the TspanC8 family are central modulators of ADAM10-mediated ectodomain shedding in vivo.
Keywords:
本文献已被 SpringerLink 等数据库收录!
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