Your browser doesn't support javascript.
loading
X-chromosome target specificity diverged between dosage compensation mechanisms of two closely related Caenorhabditis species.
Yang, Qiming; Lo, Te-Wen; Brejc, Katjusa; Schartner, Caitlin; Ralston, Edward J; Lapidus, Denise M; Meyer, Barbara J.
Affiliation
  • Yang Q; Howard Hughes Medical Institute, Berkeley, United States.
  • Lo TW; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.
  • Brejc K; Howard Hughes Medical Institute, Berkeley, United States.
  • Schartner C; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.
  • Ralston EJ; Howard Hughes Medical Institute, Berkeley, United States.
  • Lapidus DM; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.
  • Meyer BJ; Howard Hughes Medical Institute, Berkeley, United States.
Elife ; 122023 03 23.
Article in En | MEDLINE | ID: mdl-36951246
ABSTRACT
An evolutionary perspective enhances our understanding of biological mechanisms. Comparison of sex determination and X-chromosome dosage compensation mechanisms between the closely related nematode species Caenorhabditis briggsae (Cbr) and Caenorhabditis elegans (Cel) revealed that the genetic regulatory hierarchy controlling both processes is conserved, but the X-chromosome target specificity and mode of binding for the specialized condensin dosage compensation complex (DCC) controlling X expression have diverged. We identified two motifs within Cbr DCC recruitment sites that are highly enriched on X 13 bp MEX and 30 bp MEX II. Mutating either MEX or MEX II in an endogenous recruitment site with multiple copies of one or both motifs reduced binding, but only removing all motifs eliminated binding in vivo. Hence, DCC binding to Cbr recruitment sites appears additive. In contrast, DCC binding to Cel recruitment sites is synergistic mutating even one motif in vivo eliminated binding. Although all X-chromosome motifs share the sequence CAGGG, they have otherwise diverged so that a motif from one species cannot function in the other. Functional divergence was demonstrated in vivo and in vitro. A single nucleotide position in Cbr MEX can determine whether Cel DCC binds. This rapid divergence of DCC target specificity could have been an important factor in establishing reproductive isolation between nematode species and contrasts dramatically with the conservation of target specificity for X-chromosome dosage compensation across Drosophila species and for transcription factors controlling developmental processes such as body-plan specification from fruit flies to mice.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Caenorhabditis / Caenorhabditis elegans Proteins Type of study: Prognostic_studies Limits: Animals Language: En Journal: Elife Year: 2023 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Caenorhabditis / Caenorhabditis elegans Proteins Type of study: Prognostic_studies Limits: Animals Language: En Journal: Elife Year: 2023 Document type: Article