Unknown

Dataset Information

0

Membrane-type matrix metalloproteinase-3 regulates neuronal responsiveness to myelin through Nogo-66 receptor 1 cleavage.


ABSTRACT: Nogo-66 receptor 1 (NgR1) is a glycosylphosphatidylinositol-anchored receptor for myelin-associated inhibitors that restricts plasticity and axonal regrowth in the CNS. NgR1 is cleaved from the cell surface of SH-SY5Y neuroblastoma cells in a metalloproteinase-dependent manner; however, the mechanism and physiological consequence of NgR1 shedding have not been explored. We now demonstrate that NgR1 is shed from multiple populations of primary neurons. Through a loss-of-function approach, we found that membrane-type matrix metalloproteinase-3 (MT3-MMP) regulates endogenous NgR1 shedding in primary neurons. Neuronal knockdown of MT3-MMP resulted in the accumulation of NgR1 at the cell surface and reduced the accumulation of the NgR1 cleavage fragment in medium conditioned by cortical neurons. Recombinant MT1-, MT2-, MT3-, and MT5-MMPs promoted NgR1 shedding from the surface of primary neurons, and this treatment rendered neurons resistant to myelin-associated inhibitors. Introduction of a cleavage-resistant form of NgR1 reconstitutes the neuronal response to these inhibitors, demonstrating that specific metalloproteinases attenuate neuronal responses to myelin in an NgR1-dependent manner.

SUBMITTER: Ferraro GB 

PROVIDER: S-EPMC3173120 | biostudies-literature | 2011 Sep

REPOSITORIES: biostudies-literature

altmetric image

Publications

Membrane-type matrix metalloproteinase-3 regulates neuronal responsiveness to myelin through Nogo-66 receptor 1 cleavage.

Ferraro Gino B GB   Morrison Charlotte J CJ   Overall Christopher M CM   Strittmatter Stephen M SM   Fournier Alyson E AE  

The Journal of biological chemistry 20110718 36


Nogo-66 receptor 1 (NgR1) is a glycosylphosphatidylinositol-anchored receptor for myelin-associated inhibitors that restricts plasticity and axonal regrowth in the CNS. NgR1 is cleaved from the cell surface of SH-SY5Y neuroblastoma cells in a metalloproteinase-dependent manner; however, the mechanism and physiological consequence of NgR1 shedding have not been explored. We now demonstrate that NgR1 is shed from multiple populations of primary neurons. Through a loss-of-function approach, we foun  ...[more]

Similar Datasets

| S-EPMC2852886 | biostudies-literature
| S-EPMC6730300 | biostudies-literature
| S-EPMC6757674 | biostudies-literature
| S-EPMC3745902 | biostudies-literature
| S-EPMC3272441 | biostudies-literature
| S-EPMC6725623 | biostudies-literature
| S-EPMC2892845 | biostudies-literature
2012-02-28 | E-GEOD-33454 | biostudies-arrayexpress
| S-EPMC1134979 | biostudies-literature
| S-EPMC4951253 | biostudies-literature