Your browser doesn't support javascript.
loading
Carbon monoxide attenuates amyloidogenesis via down-regulation of NF-κB-mediated BACE1 gene expression.
Kim, Hyo Jeong; Joe, Yeonsoo; Chen, Yingqing; Park, Gyu Hwan; Kim, Uh-Hyun; Chung, Hun Taeg.
Affiliation
  • Kim HJ; Meta-Inflammation Research Institute of Basic Research, School of Biological Sciences, University of Ulsan, Ulsan, South Korea.
  • Joe Y; Meta-Inflammation Research Institute of Basic Research, School of Biological Sciences, University of Ulsan, Ulsan, South Korea.
  • Chen Y; Meta-Inflammation Research Institute of Basic Research, School of Biological Sciences, University of Ulsan, Ulsan, South Korea.
  • Park GH; College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea.
  • Kim UH; National Creative Research Laboratory for Ca2+ Signaling Network, Medical School, Chonbuk National University, Jeonju, South Korea.
  • Chung HT; Meta-Inflammation Research Institute of Basic Research, School of Biological Sciences, University of Ulsan, Ulsan, South Korea.
Aging Cell ; 18(1): e12864, 2019 02.
Article de En | MEDLINE | ID: mdl-30411846
ABSTRACT
Amyloid-ß (Aß) peptides, the major constituent of plaques, are generated by sequential proteolytic cleavage of the amyloid precursor protein (APP) via ß-secretase (BACE1) and the γ-secretase complex. It has been proposed that the abnormal secretion and accumulation of Aß are the initial causative events in the development of Alzheimer's disease (AD). Drugs modulating this pathway could be used for AD treatment. Previous studies indicated that carbon monoxide (CO), a product of heme oxygenase (HO)-1, protects against Aß-induced toxicity and promotes neuroprotection. However, the mechanism underlying the mitigative effect of CO on Aß levels and BACE1 expression is unclear. Here, we show that CO modulates cleavage of APP and Aß production by decreasing BACE1 expression in vivo and in vitro. CO reduces Aß levels and improves memory deficits in AD transgenic mice. The regulation of BACE1 expression by CO is dependent on nuclear factor-kappa B (NF-κB). Consistent with the negative role of SIRT1 in the NF-κB activity, CO fails to evoke significant decrease in BACE1 expression in the presence of the SIRT1 inhibitor. Furthermore, CO attenuates elevation of BACE1 level in brains of 3xTg-AD mouse model as well as mice fed high-fat, high-cholesterol diets. CO reduces the NF-κB-mediated transcription of BACE1 induced by the cholesterol oxidation product 27-hydroxycholesterol or hydrogen peroxide. These data suggest that CO reduces the NF-κB-mediated BACE1 transcription and consequently decreases Aß production. Our study provides novel mechanisms by which CO reduces BACE1 expression and Aß production and may be an effective agent for AD treatment.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Monoxyde de carbone / Régulation négative / Régulation de l'expression des gènes codant pour des enzymes / Aspartic acid endopeptidases / Facteur de transcription NF-kappa B / Amyloid precursor protein secretases / Amyloïde Type d'étude: Prognostic_studies Limites: Animals / Humans / Male Langue: En Journal: Aging Cell Année: 2019 Type de document: Article Pays d'affiliation: Corée du Sud

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Monoxyde de carbone / Régulation négative / Régulation de l'expression des gènes codant pour des enzymes / Aspartic acid endopeptidases / Facteur de transcription NF-kappa B / Amyloid precursor protein secretases / Amyloïde Type d'étude: Prognostic_studies Limites: Animals / Humans / Male Langue: En Journal: Aging Cell Année: 2019 Type de document: Article Pays d'affiliation: Corée du Sud
...