Glibenclamide-Loaded Engineered Nanovectors (GNVs) Modulate Autophagy and NLRP3-Inflammasome Activation

Author:

Saresella Marina1ORCID,Zoia Chiara Paola23ORCID,La Rosa Francesca1ORCID,Bazzini Chiara23ORCID,Sala Gessica23ORCID,Grassenis Erica23,Marventano Ivana1,Hernis Ambra1,Piancone Federica1,Conti Elisa23ORCID,Sesana Silvia4ORCID,Re Francesca4ORCID,Seneci Pierfausto5ORCID,Ferrarese Carlo236ORCID,Clerici Mario17ORCID

Affiliation:

1. IRCCS Fondazione Don Carlo Gnocchi, 20147 Milan, Italy

2. Neurobiology Laboratory, School of Medicine and Surgery, University of Study of Milano-Bicocca, 20900 Monza, Italy

3. Milan Center for Neuroscience, University of Study of Milano-Bicocca, 20126 Milano, Italy

4. BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy

5. Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milan, Italy

6. Department of Neuroscience, IRCC Fondazione S. Gerardo dei Tintori, 20900 Monza, Italy

7. Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy

Abstract

Activation of the NLRP3 inflammasome in response to either exogenous (PAMPs) or endogenous (DAMPs) stimuli results in the production of IL-18, caspase-1 and IL-1β. These cytokines have a beneficial role in promoting inflammation, but an excessive activation of the inflammasome and the consequent constitutive inflammatory status plays a role in human pathologies, including Alzheimer’s disease (AD). Autophagic removal of NLRP3 inflammasome activators can reduce inflammasome activation and inflammation. Likewise, inflammasome signaling pathways regulate autophagy, allowing the development of inflammatory responses but preventing excessive and detrimental inflammation. Nanotechnology led to the development of liposome engineered nanovectors (NVs) that can load and carry drugs. We verified in an in vitro model of AD-associated inflammation the ability of Glibenclamide-loaded NVs (GNVs) to modulate the balance between inflammasome activation and autophagy. Human THP1dM cells were LPS-primed and oligomeric Aß-stimulated in the presence/absence of GNVs. IL-1β, IL-18 and activated caspase-1 production was evaluated by the Automated Immunoassay System (ELLA); ASC speck formation (a marker of NLRP3 activation) was analyzed by FlowSight Imaging flow-cytometer (AMNIS); the expression of autophagy targets was investigated by RT-PCR and Western blot (WB); and the modulation of autophagy-related up-stream signaling pathways and Tau phosphorylation were WB-quantified. Results showed that GNVs reduce activation of the NLRP3 inflammasome and prevent the Aß-induced phosphorylation of ERK, AKT, and p70S6 kinases, potentiating autophagic flux and counteracting Tau phosphorylation. These preliminary results support the investigation of GNVs as a possible novel strategy in disease and rehabilitation to reduce inflammasome-associated inflammation.

Publisher

MDPI AG

Subject

Drug Discovery,Pharmaceutical Science,Molecular Medicine

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3