beta -Amyloid exacerbates inflammation in astrocytes lacking fatty acid amide hydrolase through a mechanism involving PPAR- alpha , PPAR-[gamma] and TRPV1, but not CB sub(1) or CB sub(2) receptors

BACKGROUND AND PURPOSE The endocannabinoid system may regulate glial cell functions and their responses to pathological stimuli, specifically, Alzheimer's disease. One experimental approach is the enhancement of endocannabinoid tone by blocking the activity of degradative enzymes, such as fatty...

Full description

Saved in:
Bibliographic Details
Published inBritish journal of pharmacology Vol. 166; no. 4; pp. 1474 - 1489
Main Authors Benito, Cristina, Tolon, Rosa Maria, Castillo, Ana Isabel, Ruiz-Valdepenas, Lourdes, Martinez-Orgado, Jose Antonio, Fernandez-Sanchez, Francisco Javier, Vazquez, Carmen, Cravatt, Benjamin F, Romero, Julian
Format Journal Article
LanguageEnglish
Published 01.06.2012
Online AccessGet full text

Cover

Loading…
More Information
Summary:BACKGROUND AND PURPOSE The endocannabinoid system may regulate glial cell functions and their responses to pathological stimuli, specifically, Alzheimer's disease. One experimental approach is the enhancement of endocannabinoid tone by blocking the activity of degradative enzymes, such as fatty acid amide hydrolase (FAAH). EXPERIMENTAL APPROACH We examined the role of FAAH in the response of astrocytes to the pathologic form of beta -amyloid (A beta ). Astrocytes from wild-type mice (WT) and from mice lacking FAAH (FAAH-KO) were incubated with A beta for 8, 24 and 48h, and their inflammatory responses were quantified by elisa, western-blotting and real-time quantitative-PCR. KEY RESULTS FAAH-KO astrocytes were significantly more responsive to A beta than WT astrocytes, as shown by the higher production of pro-inflammatory cytokines. Expression of COX-2, inducible NOS and TNF- alpha was also increased in A beta -exposed KO astrocytes compared with that in WTs. These effects were accompanied by a differential pattern of activation of signalling cascades involved in mediating inflammatory responses, such as ERK1/2, p38MAPK and NF[kappa] B. PPAR- alpha and PPAR-[gamma] as well as transient receptor potential vanilloid-1 (TRPV1), but not cannabinoid CB sub(1) or CB sub(2) receptors, mediate some of the differential changes observed in A beta -exposed FAAH-KO astrocytes. The pharmacological blockade of FAAH did not render astrocytes more sensitive to A beta . In contrast, exogenous addition of several acylethanolamides (anandamide, palmitoylethanolamide and oleoylethanolamide) induced an antiinflammatory response. CONCLUSIONS The genetic deletion of FAAH in astrocytes exacerbated their inflammatory phenotype against A beta in a process involving PPAR- alpha , PPAR-[gamma] and TRPV1 receptors.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
content type line 23
ObjectType-Feature-2
ISSN:0007-1188
1476-5381
DOI:10.1111/j.1476-5381.2012.01889.x