Abstract

We demonstrate how RNA binding protein FOX-1 functions as a dose-dependent X-signal element to communicate X-chromosome number and thereby determine nematode sex. FOX-1, an RNA recognition motif protein, triggers hermaphrodite development in XX embryos by causing non-productive alternative pre-mRNA splicing of xol-1, the master sex-determination switch gene that triggers male development in XO embryos. RNA binding experiments together with genome editing demonstrate that FOX-1 binds to multiple GCAUG and GCACG motifs in a xol-1 intron, causing intron retention or partial exon deletion, thereby eliminating male-determining XOL-1 protein. Transforming all motifs to GCAUG or GCACG permits accurate alternative splicing, demonstrating efficacy of both motifs. Mutating subsets of both motifs partially alleviates non-productive splicing. Mutating all motifs blocks it, as does transforming them to low-affinity GCUUG motifs. Combining multiple high-affinity binding sites with the twofold change in FOX-1 concentration between XX and XO embryos achieves dose-sensitivity in splicing regulation to determine sex.

Highlights

  • Determining sex is one of the most fundamental developmental decisions that most organisms must make

  • Our prior experiments showed that overexpression of FOX-1 by itself is sufficient to repress endogenous xol-1 activity, causing XO embryos to adopt the hermaphrodite sexual fate and die from reduced X-chromosome expression triggered by binding of the dosage compensation complex (DCC) to the single X (Nicoll et al, 1997)

  • The xol-1(+) transgenes were expressed in XX animals at a somewhat higher level than the endogenous xol-1 gene: the seven XX array lines could only be maintained if both endogenous copies of fox-1 were wild type

Read more

Summary

Introduction

Determining sex is one of the most fundamental developmental decisions that most organisms must make. The X:A signal determines sex by controlling the activity of its direct target, the master sex-determination switch gene xol-1 (XO lethal) (Figure 1; Carmi et al, 1998; Farboud et al, 2013; Meyer, 2018; Miller et al, 1988; Nicoll et al, 1997; Powell et al, 2005; Rhind et al, 1995). Xol-1 controls the choice of sexual fate and X-chromosome gene expression through the process of X-chromosome dosage compensation (Meyer, 2018; Miller et al, 1988; Rhind et al, 1995).

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.