Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 14: 1171937, 2023.
Article in English | MEDLINE | ID: mdl-37533630

ABSTRACT

High levels of plasma cholesterol, especially high levels of low-density lipoprotein cholesterol (LDL-C), have been associated with an increased risk of Alzheimer's disease. The cholesteryl ester transfer protein (CETP) in plasma distributes cholesteryl esters between lipoproteins and increases LDL-C in plasma. Epidemiologically, decreased CETP activity has been associated with sustained cognitive performance during aging, longevity, and a lower risk of Alzheimer's disease. Thus, pharmacological CETP inhibitors could be repurposed for the treatment of Alzheimer's disease as they are safe and effective at lowering CETP activity and LDL-C. Although CETP is mostly expressed by the liver and secreted into the bloodstream, it is also expressed by astrocytes in the brain. Therefore, it is important to determine whether CETP inhibitors can enter the brain. Here, we describe the pharmacokinetic parameters of the CETP inhibitor evacetrapib in the plasma, liver, and brain tissues of CETP transgenic mice. We show that evacetrapib crosses the blood-brain barrier and is detectable in brain tissue 0.5 h after a 40 mg/kg i.v. injection in a non-linear function. We conclude that evacetrapib may prove to be a good candidate to treat CETP-mediated cholesterol dysregulation in Alzheimer's disease.

2.
J Lipid Res ; 63(9): 100260, 2022 09.
Article in English | MEDLINE | ID: mdl-35921880

ABSTRACT

The cholesteryl ester transfer protein (CETP) is a lipid transfer protein responsible for the exchange of cholesteryl esters and triglycerides between lipoproteins. Decreased CETP activity is associated with longevity, cardiovascular health, and maintenance of good cognitive performance. Interestingly, mice lack the CETP-encoding gene and have very low levels of LDL particles compared with humans. Currently, the molecular mechanisms induced because of CETP activity are not clear. To understand how CETP activity affects the brain, we utilized CETP transgenic (CETPtg) mice that show elevated LDL levels upon induction of CETP expression through a high-cholesterol diet. CETPtg mice on a high-cholesterol diet showed up to 22% higher cholesterol levels in the brain. Using a microarray on mostly astrocyte-derived mRNA, we found that this cholesterol increase is likely not because of elevated de novo synthesis of cholesterol. However, cholesterol efflux is decreased in CETPtg mice along with an upregulation of the complement factor C1Q, which plays a role in neuronal cholesterol clearance. Our data suggest that CETP activity affects brain health through modulating cholesterol distribution and clearance. Therefore, we propose that CETPtg mice constitute a valuable research tool to investigate the impact of cholesterol metabolism on brain function.


Subject(s)
Hypercholesterolemia , Hyperlipidemias , Animals , Brain/metabolism , Cholesterol/metabolism , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Esters/metabolism , Complement C1q/metabolism , Humans , Hypercholesterolemia/metabolism , Hyperlipidemias/metabolism , Lipoproteins/metabolism , Liver/metabolism , Mice , RNA, Messenger/genetics , Triglycerides/metabolism
3.
J Membr Biol ; 251(3): 369-378, 2018 06.
Article in English | MEDLINE | ID: mdl-29260282

ABSTRACT

Proteases, sharp yet unforgivable tools of every cell, require tight regulation to ensure specific non-aberrant cleavages. The relatively recent discovered class of intramembrane proteases has gained increasing interest due to their involvement in important signaling pathways linking them to diseases including Alzheimer's disease and cancer. Despite tremendous efforts, their regulatory mechanisms have only started to unravel. There is evidence that the membrane composition itself can regulate intramembrane protease activity and specificity. In this review, we highlight the work on γ-secretase and rhomboid proteases and summarize several studies as to how different lipids impact on enzymatic activity.


Subject(s)
Cell Membrane/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Endopeptidases/metabolism , Membrane Proteins/genetics , Protein Binding , Proteolysis , Substrate Specificity
4.
J Biol Chem ; 291(42): 21903-21912, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27563067

ABSTRACT

The amyloid precursor protein (APP) is an ubiquitously expressed cell surface protein and a key molecule in the etiology of Alzheimer disease. Amyloidogenic processing of APP through secretases leads to the generation of toxic amyloid ß (Aß) peptides, which are regarded as the molecular cause of the disease. We report here an alternative processing pathway of APP through the mammalian intramembrane rhomboid protease RHBDL4. RHBDL4 efficiently cleaves APP inside the cell, thus bypassing APP from amyloidogenic processing, leading to reduced Aß levels. RHBDL4 cleaves APP multiple times in the ectodomain, resulting in several N- and C-terminal fragments that are not further degraded by classical APP secretases. Knockdown of endogenous RHBDL4 results in decreased levels of C-terminal fragments derived from endogenous APP. Similarly, we found the APP family members APLP1 and APLP2 to be substrates of RHBDL4. We conclude that RHBDL4-mediated APP processing provides insight into APP and rhomboid physiology and qualifies for further investigations to elaborate its impact on Alzheimer disease pathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Membrane Proteins/metabolism , Protein Processing, Post-Translational , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cell Line , Humans , Membrane Proteins/genetics , Protein Domains
5.
Biochemistry ; 54(17): 2777-84, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25875527

ABSTRACT

Amyloid-ß (Aß) peptides are likely the molecular cause of neurodegeneration observed in Alzheimer's disease. In the brain, Aß42 and Aß40 are toxic and the most important proteolytic fragments generated through sequential processing of the amyloid precursor protein (APP) by ß- and γ-secretases. Impeding the generation of Aß42 and Aß40 is thus considered as a promising strategy to prevent Alzheimer's disease. We therefore wanted to determine key parameters of the APP transmembrane sequence enabling production of these Aß species. Here we show that the hydrophilicity of amino acid residues G33, T43, and T48 critically determines the generation of Aß42 and Aß40 peptides (amino acid numbering according to Aß nomenclature starting with aspartic acid 1). First, we performed a comprehensive mutational analysis of glycine residue G33 positioned within the N-terminal half of the APP transmembrane sequence by exchanging it against the 19 other amino acids. We found that hydrophilicity of the residue at position 33 positively correlated with Aß42 and Aß40 generation. Second, we analyzed two threonine residues at positions T43 and T48 in the C-terminal half of the APP-transmembrane sequence. Replacement of single threonine residues by hydrophobic valines inversely affected Aß42 and Aß40 generation. We observed that threonine mutants affected the initial γ-secretase cut, which is associated with levels of Aß42 or Aß40. Overall, hydrophilic residues of the APP transmembrane sequence decide on the exact initial γ-cut and the amounts of Aß42 and Aß40.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Amyloid beta-Protein Precursor/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Blotting, Western , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...