Abstract

To determine the presence of sexual dimorphic transcription and how in vitro culture environments influence X-linked gene transcription patterns in preimplantation embryos, we analyzed mRNA expression levels in in vivo-derived, in vitro-fertilized (IVF), and cloned porcine blastocysts. Our results clearly show that sex-biased expression occurred between female and male in vivo blastocysts in X-linked genes. The expression levels of XIST, G6PD, HPRT1, PGK1, and BEX1 were significantly higher in female than in male blastocysts, but ZXDA displayed higher levels in male than in female blastocysts. Although we found aberrant expression patterns for several genes in IVF and cloned blastocysts, similar sex-biased expression patterns (on average) were observed between the sexes. The transcript levels of BEX1 and XIST were upregulated and PGK1 was downregulated in both IVF and cloned blastocysts compared with in vivo counterparts. Moreover, a remarkable degree of expression heterogeneity was observed among individual cloned embryos (the level of heterogeneity was similar in both sexes) but only a small proportion of female IVF embryos exhibited variability, indicating that this phenomenon may be primarily caused by faulty reprogramming by the somatic cell nuclear transfer (SCNT) process rather than in vitro conditions. Aberrant expression patterns in cloned embryos of both sexes were not ameliorated by treatment with Scriptaid as a potent HDACi, although the blastocyst rate increased remarkably after this treatment. Taken together, these results indicate that female and male porcine blastocysts produced in vivo and in vitro transcriptional sexual dimorphisms in the selected X-linked genes and compensation of X-linked gene dosage may not occur at the blastocyst stage. Moreover, altered X-linked gene expression frequently occurred in porcine IVF and cloned embryos, indicating that X-linked gene regulation is susceptible to in vitro culture and the SCNT process, which may eventually lead to problems with embryonic or placental defects.

Highlights

  • The onset of X chromosome inactivation (XCI) is regulated by the X-inactive specific transcript (Xist), which coats the inactive X chromosome in cis and compensates for sex-linked gene dosage differences

  • The expression levels of brain expressed X-linked protein 1 (BEX1), G6PD, HPRT1, PGK1, and XIST were significantly higher in female than in male blastocysts (P,0.01), while ZXDA levels were significantly higher in males than in females (P = 0.0002)

  • These results indicate that porcine embryos showed transcriptional sexual dimorphisms in X-linked genes at the blastocyst stage

Read more

Summary

Introduction

The onset of X chromosome inactivation (XCI) is regulated by the X-inactive specific transcript (Xist), which coats the inactive X chromosome in cis and compensates for sex-linked gene dosage differences. Evidence from global transcriptome analysis indicates that a large number of X-linked genes are regulated differently between the sexes in early mammalian embryos [4,5] This transcriptional level sexual dimorphism creates differences in developmental kinetics and epigenetics between female and male embryos during preimplantation development [6]. Sexual dimorphisms in gene expression are found in the placenta between; female and male conceptuses respond differently to diet changes in the maternal environment [7]. These differences can lead to sexspecific embryo viability under in vitro conditions, such as cryopreservation and stressful culture systems (i.e., nutritional or oxidative conditions), and sex-ratio skewing of offspring because of environmental factors [8,9,10]

Methods
Results
Conclusion
Full Text
Published version (Free)

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