Aberrant R-loop-induced replication stress in MED12-mutant uterine fibroids

S Muralimanoharan, R Shamby, N Stansbury… - Scientific reports, 2022 - nature.com
S Muralimanoharan, R Shamby, N Stansbury, R Schenken, B de la Pena Avalos
Scientific reports, 2022nature.com
Uterine fibroid (UF) driver mutations in Mediator complex subunit 12 (MED12) trigger
genomic instability and tumor development through unknown mechanisms. Herein, we show
that MED12 mutations trigger aberrant R-loop-induced replication stress, suggesting a
possible route to genomic instability and a novel therapeutic vulnerability in this dominant
UF subclass. Immunohistochemical analyses of patient-matched tissue samples revealed
that MED12 mutation-positive UFs, compared to MED12 mutation-negative UFs and …
Abstract
Uterine fibroid (UF) driver mutations in Mediator complex subunit 12 (MED12) trigger genomic instability and tumor development through unknown mechanisms. Herein, we show that MED12 mutations trigger aberrant R-loop-induced replication stress, suggesting a possible route to genomic instability and a novel therapeutic vulnerability in this dominant UF subclass. Immunohistochemical analyses of patient-matched tissue samples revealed that MED12 mutation-positive UFs, compared to MED12 mutation-negative UFs and myometrium, exhibited significantly higher levels of R-loops and activated markers of Ataxia Telangiectasia and Rad3-related (ATR) kinase-dependent replication stress signaling in situ. Single molecule DNA fiber analysis revealed that primary cells from MED12 mutation-positive UFs, compared to those from patient-matched MED12 mutation-negative UFs and myometrium, exhibited defects in replication fork dynamics, including reduced fork speeds, increased and decreased numbers of stalled and restarted forks, respectively, and increased asymmetrical bidirectional forks. Notably, these phenotypes were recapitulated and functionally linked in cultured uterine smooth muscle cells following chemical inhibition of Mediator-associated CDK8/19 kinase activity that is known to be disrupted by UF driver mutations in MED12. Thus, Mediator kinase inhibition triggered enhanced R-loop formation and replication stress leading to an S-phase cell cycle delay, phenotypes that were rescued by overexpression of the R-loop resolving enzyme RNaseH. Altogether, these findings reveal MED12-mutant UFs to be uniquely characterized by aberrant R-loop induced replication stress, suggesting a possible basis for genomic instability and new avenues for therapeutic intervention that involve the replication stress phenotype in this dominant UF subtype.
nature.com