4.6 Article

Cerebral angiogenesis ameliorates pathological disorders in Nemo-deficient mice with small-vessel disease

Journal

JOURNAL OF CEREBRAL BLOOD FLOW AND METABOLISM
Volume 41, Issue 2, Pages 219-235

Publisher

SAGE PUBLICATIONS INC
DOI: 10.1177/0271678X20910522

Keywords

Angiogenesis; small vessel disease; incontinentia pigmenti; NF-kappa B; endothelial cell

Funding

  1. Deutsche Forschungsgemeinschaft [SCHW 416/5-2, GRK 1957]
  2. European Union [607962]
  3. European Research Council (ERC) under the European Union [810331]

Ask authors/readers for more resources

The study investigated angiogenesis in cerebral small-vessel diseases, revealing the crucial role of Nemo in productive angiogenesis and its potential to counterbalance vessel loss. Experimental results showed that newborn endothelial cells were particularly vulnerable to ongoing recombination.
Cerebral small-vessel diseases (SVDs) often follow a progressive course. Little is known about the function of angiogenesis, which potentially induces regression of SVDs. Here, we investigated angiogenesis in a mouse model of incontinentia pigmenti (IP), a genetic disease comprising features of SVD. IP is caused by inactivating mutations of Nemo, the essential component of NF-kappa B signaling. When deleting Nemo in the majority of brain endothelial cells (Nemo(beKO) mice), the transcriptional profile of vessels indicated cell proliferation. Brain endothelial cells expressed Ki67 and showed signs of DNA synthesis. In addition to cell proliferation, we observed sprouting and intussusceptive angiogenesis in Nemo(beKO) mice. Angiogenesis occurred in all segments of the vasculature and in proximity to vessel rarefaction and tissue hypoxia. Apparently, NEMO was required for productive angiogenesis because endothelial cells that had escaped Nemo inactivation showed a higher proliferation rate than Nemo-deficient cells. Therefore, newborn endothelial cells were particularly vulnerable to ongoing recombination. When we interfered with productive angiogenesis by inducing ongoing ablation of Nemo, mice did not recover from IP manifestations but rather showed severe functional deficits. In summary, the data demonstrate that angiogenesis is present in this model of SVD and suggest that it may counterbalance the loss of vessels.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available