In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15

Standard

In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15. / Seipold, Lisa; Altmeppen, Hermann; Koudelka, Tomas; Tholey, Andreas; Kasparek, Petr; Sedlacek, Radislav; Schweizer, Michaela; Bär, Julia; Mikhaylova, Marina; Glatzel, Markus; Saftig, Paul.

In: CELL MOL LIFE SCI, Vol. 75, No. 17, 09.2018, p. 3251-3267.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Seipold, L, Altmeppen, H, Koudelka, T, Tholey, A, Kasparek, P, Sedlacek, R, Schweizer, M, Bär, J, Mikhaylova, M, Glatzel, M & Saftig, P 2018, 'In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15', CELL MOL LIFE SCI, vol. 75, no. 17, pp. 3251-3267. https://doi.org/10.1007/s00018-018-2791-2

APA

Seipold, L., Altmeppen, H., Koudelka, T., Tholey, A., Kasparek, P., Sedlacek, R., Schweizer, M., Bär, J., Mikhaylova, M., Glatzel, M., & Saftig, P. (2018). In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15. CELL MOL LIFE SCI, 75(17), 3251-3267. https://doi.org/10.1007/s00018-018-2791-2

Vancouver

Bibtex

@article{f0424a15001841f2a53ebd8d7779519e,
title = "In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15",
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 = "Journal Article",
author = "Lisa Seipold and Hermann Altmeppen and Tomas Koudelka and Andreas Tholey and Petr Kasparek and Radislav Sedlacek and Michaela Schweizer and Julia B{\"a}r and Marina Mikhaylova and Markus Glatzel and Paul Saftig",
year = "2018",
month = sep,
doi = "10.1007/s00018-018-2791-2",
language = "English",
volume = "75",
pages = "3251--3267",
journal = "CELL MOL LIFE SCI",
issn = "1420-682X",
publisher = "Birkhauser Verlag Basel",
number = "17",

}

RIS

TY - JOUR

T1 - In vivo regulation of the A disintegrin and metalloproteinase 10 (ADAM10) by the tetraspanin 15

AU - Seipold, Lisa

AU - Altmeppen, Hermann

AU - Koudelka, Tomas

AU - Tholey, Andreas

AU - Kasparek, Petr

AU - Sedlacek, Radislav

AU - Schweizer, Michaela

AU - Bär, Julia

AU - Mikhaylova, Marina

AU - Glatzel, Markus

AU - Saftig, Paul

PY - 2018/9

Y1 - 2018/9

N2 - 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.

AB - 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.

KW - Journal Article

U2 - 10.1007/s00018-018-2791-2

DO - 10.1007/s00018-018-2791-2

M3 - SCORING: Journal article

C2 - 29520422

VL - 75

SP - 3251

EP - 3267

JO - CELL MOL LIFE SCI

JF - CELL MOL LIFE SCI

SN - 1420-682X

IS - 17

ER -