Characterization of the molecular mechanisms that govern anti‐Müllerian hormone synthesis and activity

Author:

Stocker William A.1ORCID,Howard James A.2ORCID,Maskey Shreya1,Luan Haitong1,Harrison Sophie G.1ORCID,Hart Kaitlin N.2ORCID,Hok Lucija3ORCID,Thompson Thomas B.3ORCID,Walton Kelly L.14ORCID,Harrison Craig A.1ORCID

Affiliation:

1. Department of Physiology, Monash Biomedicine Discovery Institute Monash University Clayton Victoria Australia

2. Department of Pharmacology and Systems Physiology University of Cincinnati Cincinnati Ohio USA

3. Department of Molecular and Cellular Biosciences University of Cincinnati Cincinnati Ohio USA

4. School of Biomedical Sciences The University of Queensland Brisbane Queensland Australia

Abstract

AbstractThe roles of anti‐Müllerian hormone (AMH) continue to expand, from its discovery as a critical factor in sex determination, through its identification as a regulator of ovarian folliculogenesis, its use in fertility clinics as a measure of ovarian reserve, and its emerging role in hypothalamic–pituitary function. In light of these actions, AMH is considered an attractive therapeutic target to address diverse reproductive needs, including fertility preservation. Here, we set out to characterize the molecular mechanisms that govern AMH synthesis and activity. First, we enhanced the processing of the AMH precursor to >90% by introducing more efficient proprotein convertase cleavage sites (RKKR or ISSRKKRSVSS [SCUT]). Importantly, enhanced processing corresponded with a dramatic increase in secreted AMH activity. Next, based on species differences across the AMH type II receptor‐binding interface, we generated a series of human AMH variants and assessed bioactivity. AMHSCUT potency (EC50 4 ng/mL) was increased 5‐ or 10‐fold by incorporating Gln484Met/Leu535Thr (EC50 0.8 ng/mL) or Gln484Met/Gly533Ser (EC50 0.4 ng/mL) mutations, respectively. Furthermore, the Gln484Met/Leu535Thr double mutant displayed enhanced efficacy, relative to AMHSCUT. Finally, we identified residues within the wrist pre‐helix of AMH (Trp494, Gln496, Ser497, and Asp498) that likely mediate type I receptor binding. Mutagenesis of these residues generated gain‐ (Trp494Phe or Gln496Leu) or loss‐ (Ser497Ala) of function AMH variants. Surprisingly, combining activating type I and type II receptor mutations only led to modest additive increases in AMH potency/efficacy. Our study is the first to characterize AMH residues involved in type I receptor binding and suggests a step‐wise receptor‐complex assembly mechanism, in which enhancement in the affinity of the ligand for either receptor can increase AMH activity beyond the natural level.

Funder

National Health and Medical Research Council

Publisher

Wiley

Subject

Genetics,Molecular Biology,Biochemistry,Biotechnology

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