4.8 Article

Chemical and genetic rescue of in vivo progranulin-deficient lysosomal and autophagic defects

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.2022115118

Keywords

frontotemporal dementia; progranulin; Caenorhabditis elegans

Funding

  1. NIH Office of Research Infrastructure Programs [P40 OD010440]
  2. National Bioresource Project for the nematode (Japan)

Ask authors/readers for more resources

The article discusses the link between GRN mutations and frontotemporal dementia (FTD) discovered in 2006 and highlights the need for advancing genetic and small-molecule therapeutics for GRN-related FTD. Research using the nematode model, Caenorhabditis elegans, shows that loss of nematode GRN ortholog results in behavioral and molecular defects, and implicates the sphingolipid metabolic pathway in regulating these defects. High-throughput drug screening using nematodes has identified two small molecules with potential therapeutic applications against GRN/pgrn-1 deficiency, offering avenues for mechanistic and therapeutic research into GRN-related neurodegeneration.
In 2006, GRN mutations were first linked to frontotemporal dementia (FTD), the leading cause of non-Alzheimer dementias. While much research has been dedicated to understanding the genetic causes of the disease, our understanding of the mechanistic impacts of GRN deficiency has only recently begun to take shape. With no known cure or treatment available for GRN-related FTD, there is a growing need to rapidly advance genetic and/or smallmolecule therapeutics for this disease. This issue is complicated by the fact that, while lysosomal dysfunction seems to be a key driver of pathology, the mechanisms linking a loss of GRN to a pathogenic state remain unclear. In our attempt to address these key issues, we have turned to the nematode, Caenorhabditis elegans, to model, study, and find potential therapies for GRN-deficient FTD. First, we show that the loss of the nematode GRN ortholog, pgrn-1, results in several behavioral and molecular defects, including lysosomal dysfunction and defects in autophagic flux. Our investigations implicate the sphingolipid metabolic pathway in the regulation of many of the in vivo defects associated with pgrn-1 loss. Finally, we utilized these nematodes as an in vivo tool for high-throughput drug screening and identified two small molecules with potential therapeutic applications against GRN/pgrn-1 deficiency. These compounds reverse the biochemical, cellular, and functional phenotypes of GRN deficiency. Together, our results open avenues for mechanistic and therapeutic research into the outcomes of GRN-related neurodegeneration, both genetic and molecular.

Authors

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

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available