Your browser doesn't support javascript.
loading
Characterization of the molecular mechanisms that govern anti-Müllerian hormone synthesis and activity.
Stocker, William A; Howard, James A; Maskey, Shreya; Luan, Haitong; Harrison, Sophie G; Hart, Kaitlin N; Hok, Lucija; Thompson, Thomas B; Walton, Kelly L; Harrison, Craig A.
Affiliation
  • Stocker WA; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
  • Howard JA; Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA.
  • Maskey S; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
  • Luan H; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
  • Harrison SG; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
  • Hart KN; Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA.
  • Hok L; Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, Ohio, USA.
  • Thompson TB; Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, Ohio, USA.
  • Walton KL; Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
  • Harrison CA; School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia.
FASEB J ; 38(1): e23377, 2024 01.
Article in En | MEDLINE | ID: mdl-38133902
ABSTRACT
The 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 Gln484 Met/Leu535 Thr (EC50 0.8 ng/mL) or Gln484 Met/Gly533 Ser (EC50 0.4 ng/mL) mutations, respectively. Furthermore, the Gln484 Met/Leu535 Thr 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- (Trp494 Phe or Gln496 Leu) or loss- (Ser497 Ala) 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.
Subject(s)
Key words

Full text: 1 Database: MEDLINE Main subject: Peptide Hormones / Anti-Mullerian Hormone Limits: Female / Humans Language: En Year: 2024 Type: Article

Full text: 1 Database: MEDLINE Main subject: Peptide Hormones / Anti-Mullerian Hormone Limits: Female / Humans Language: En Year: 2024 Type: Article