Erythrocyte ENT1-AMPD3 Axis is an Essential Purinergic Hypoxia Sensor and Energy Regulator Combating CKD in a Mouse Model

Author:

Chen Changhan1234,Xie TingTing145ORCID,Zhang Yujin1,Wang Yiyan1234ORCID,Yu Fang16,Lin Lizhen17ORCID,Zhang Weiru45,Brown Benjamin C.7ORCID,Zhang Xin234,Kellems Rodney E.8,D'Alessandro Angelo9ORCID,Xia Yang1234ORCID

Affiliation:

1. National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China

2. Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China

3. Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, China

4. National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China

5. Department of General Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China

6. Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China

7. Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China

8. Department of Biochemistry and Molecular Biology, The University of Texas McGovern Medical School at Houston, Houston, Texas

9. Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado

Abstract

Significance Statement Hypoxia drives kidney damage and progression of CKD. Although erythrocytes respond rapidly to hypoxia, their role and the specific molecules sensing and responding to hypoxia in CKD remain unclear. In this study, we demonstrated in a mouse model that erythrocyte ENT1-AMPD3 is a master energy regulator of the intracellular purinergic hypoxic compensatory response that promotes rapid energy supply from extracellular adenosine, eAMPK-dependent metabolic reprogramming, and O2 delivery, which combat renal hypoxia and progression of CKD. ENT1-AMPD3-AMPK-BPGM comprise a group of circulating erythroid-specific biomarkers, providing early diagnostic and novel therapeutic targets for CKD. Background Hypoxia drives kidney damage and progression of CKD. Although erythrocytes respond rapidly to hypoxia, their role and the specific molecules sensing and responding to hypoxia in CKD remain unclear. Methods Mice with an erythrocyte-specific deficiency in equilibrative nucleoside transporter 1 (eEnt1 −/− ) and a global deficiency in AMP deaminase 3 (Ampd3 −/− ) were generated to define their function in two independent CKD models, including angiotensin II (Ang II) infusion and unilateral ureteral obstruction (UUO). Unbiased metabolomics, isotopic adenosine flux, and various biochemical and cell culture analyses coupled with genetic studies were performed. Translational studies in patients with CKD and cultured human erythrocytes examined the role of ENT1 and AMPD3 in erythrocyte function and metabolism. Results eEnt1 −/− mice display severe renal hypoxia, kidney damage, and fibrosis in both CKD models. The loss of eENT1-mediated adenosine uptake reduces intracellular AMP and thus abolishes the activation of AMPKα and bisphosphoglycerate mutase (BPGM). This results in reduced 2,3-bisphosphoglycerate and glutathione, leading to overwhelming oxidative stress in eEnt1 −/− mice. Excess reactive oxygen species (ROS) activates AMPD3, resulting in metabolic reprogramming and reduced O2 delivery, leading to severe renal hypoxia in eEnt1 −/− mice. By contrast, genetic ablation of AMPD3 preserves the erythrocyte adenine nucleotide pool, inducing AMPK-BPGM activation, O2 delivery, and antioxidative stress capacity, which protect against Ang II-induced renal hypoxia, damage, and CKD progression. Translational studies recapitulated the findings in mice. Conclusion eENT1-AMPD3, two highly enriched erythrocyte purinergic components that sense hypoxia, promote eAMPK-BPGM–dependent metabolic reprogramming, O2 delivery, energy supply, and antioxidative stress capacity, which mitigates renal hypoxia and CKD progression.

Publisher

Ovid Technologies (Wolters Kluwer Health)

Subject

Nephrology,General Medicine

Reference56 articles.

1. Prevalence and disease burden of chronic kidney disease;Lv;Adv Exp Med Biol.,2019

2. Alterations of the human gut microbiome in chronic kidney disease;Ren;Adv Sci (Weinh).,2020

3. Chronic kidney disease;Kalantar-Zadeh;Lancet.,2021

4. Non-invasive molecular imaging of kidney diseases;Klinkhammer;Nat Rev Nephrol.,2021

5. Progressive renal disease: the chronic hypoxia hypothesis;Fine;Kidney Int Suppl.,1998

Cited by 1 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