Angiotensin II–Mediated Neuroinflammation in the Hippocampus Contributes to Neuronal Deficits and Cognitive Impairment in Heart Failure Rats

Author:

Althammer Ferdinand12,Roy Ranjan K.1ORCID,Kirchner Matthew K.1ORCID,Campos-Lira Elba13,Whitley Kathryn E.3ORCID,Davis Steven3,Montanez Juliana1,Ferreira-Neto Hildebrando Candido1,Danh Jessica4ORCID,Feresin Rafaela4ORCID,Biancardi Vinicia Campana5ORCID,Zafar Usama13,Parent Marise B.136ORCID,Stern Javier E.13ORCID

Affiliation:

1. Center for Neuroinflammation and Cardiometabolic Diseases (F.A., R.K.R., M.K.K., E.C.-L., J.M., H.C.F.-N., U.Z., M.B.P., J.E.S.), Georgia State University, Atlanta.

2. Now with Institute of Human Genetics, University Hospital Heidelberg (F.A.).

3. Neuroscience Institute (E.C.-L., K.E.W., S.D., U.Z., M.B.P., J.E.S.), Georgia State University, Atlanta.

4. Department of Nutrition (J.D., R.F.), Georgia State University, Atlanta.

5. Anatomy, Physiology, & Pharmacology, College of Veterinary Medicine, Auburn University, AL (V.C.B.).

6. Department of Psychology (M.B.P.), Georgia State University, Atlanta.

Abstract

Background: Heart failure (HF) is a debilitating disease affecting >64 million people worldwide. In addition to impaired cardiovascular performance and associated systemic complications, most patients with HF suffer from depression and substantial cognitive decline. Although neuroinflammation and brain hypoperfusion occur in humans and rodents with HF, the underlying neuronal substrates, mechanisms, and their relative contribution to cognitive deficits in HF remains unknown. Methods: To address this critical gap in our knowledge, we used a well-established HF rat model that mimics clinical outcomes observed in the human population, along with a multidisciplinary approach combining behavioral, electrophysiological, neuroanatomical, molecular and systemic physiological approaches. Results: Our studies support neuroinflammation, hypoperfusion/hypoxia, and neuronal deficits in the hippocampus of HF rats, which correlated with the progression and severity of the disease. An increased expression of AT1aRs (Ang II [angiotensin II] receptor type 1a) in hippocampal microglia preceded the onset of neuroinflammation. Importantly, blockade of AT1Rs with a clinically used therapeutic drug (Losartan), and delivered in a clinically relevant manner, efficiently reversed neuroinflammatory end points (but not hypoxia ones), resulting in turn in improved cognitive performance in HF rats. Finally, we show than circulating Ang II can leak and access the hippocampal parenchyma in HF rats, constituting a possible source of Ang II initiating the neuroinflammatory signaling cascade in HF. Conclusions: In this study, we identified a neuronal substrate (hippocampus), a mechanism (Ang II–driven neuroinflammation) and a potential neuroprotective therapeutic target (AT1aRs) for the treatment of cognitive deficits in HF.

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Internal Medicine

Cited by 2 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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