Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34083437

ABSTRACT

Transposable elements (TEs) are mobile sequences that engender widespread mutations and thus are a major hazard that must be silenced. The most abundant active class of TEs in mammalian genomes is long interspersed element class 1 (LINE1). Here, we report that LINE1 transposition is suppressed in the male germline by transcription factors encoded by a rapidly evolving X-linked homeobox gene cluster. LINE1 transposition is repressed by many members of this RHOX transcription factor family, including those with different patterns of expression during spermatogenesis. One family member-RHOX10-suppresses LINE1 transposition during fetal development in vivo when the germline would otherwise be susceptible to LINE1 activation because of epigenetic reprogramming. We provide evidence that RHOX10 suppresses LINE transposition by inducing Piwil2, which encodes a key component in the Piwi-interacting RNA pathway that protects against TEs. The ability of RHOX transcription factors to suppress LINE1 is conserved in humans but is lost in RHOXF2 mutants from several infertile human patients, raising the possibility that loss of RHOXF2 causes human infertility by allowing uncontrolled LINE1 expression in the germline. Together, our results support a model in which the Rhox gene cluster is in an evolutionary arms race with TEs, resulting in expansion of the Rhox gene cluster to suppress TEs in different biological contexts.


Subject(s)
DNA Transposable Elements/genetics , Germ Cells/metabolism , Long Interspersed Nucleotide Elements/genetics , Long Interspersed Nucleotide Elements/physiology , Multigene Family , Animals , Gene Expression Regulation , Genes, X-Linked , HEK293 Cells , Homeodomain Proteins , Humans , Male , Mice , Mice, Inbred C57BL , Spermatogenesis/genetics , Transcription Factors/metabolism
2.
Dev Cell ; 44(3): 392-402.e7, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29408237

ABSTRACT

Global transcriptional silencing is a highly conserved mechanism central to the oocyte-to-embryo transition. We report the unexpected discovery that global transcriptional silencing in oocytes depends on an mRNA decay activator. Oocyte-specific loss of ZFP36L2 an RNA-binding protein that promotes AU-rich element-dependent mRNA decay prevents global transcriptional silencing and causes oocyte maturation and fertilization defects, as well as complete female infertility in the mouse. Single-cell RNA sequencing revealed that ZFP36L2 downregulates mRNAs encoding transcription and chromatin modification regulators, including a large group of mRNAs for histone demethylases targeting H3K4 and H3K9, which we show are bound and degraded by ZFP36L2. Oocytes lacking Zfp36l2 fail to accumulate histone methylation at H3K4 and H3K9, marks associated with the transcriptionally silent, developmentally competent oocyte state. Our results uncover a ZFP36L2-dependent mRNA decay mechanism that acts as a developmental switch during oocyte growth, triggering wide-spread shifts in chromatin modification and global transcription.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Infertility, Female/pathology , Oocytes/metabolism , Transcription, Genetic , Tristetraprolin/physiology , Animals , Female , High-Throughput Nucleotide Sequencing , Infertility, Female/genetics , Infertility, Female/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oogenesis/genetics , RNA Stability/genetics , Single-Cell Analysis , Transcriptome
3.
Cell Rep ; 17(1): 149-164, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27681428

ABSTRACT

The developmental origins of most adult stem cells are poorly understood. Here, we report the identification of a transcription factor-RHOX10-critical for the initial establishment of spermatogonial stem cells (SSCs). Conditional loss of the entire 33-gene X-linked homeobox gene cluster that includes Rhox10 causes progressive spermatogenic decline, a phenotype indistinguishable from that caused by loss of only Rhox10. We demonstrate that this phenotype results from dramatically reduced SSC generation. By using a battery of approaches, including single-cell-RNA sequencing (scRNA-seq) analysis, we show that Rhox10 drives SSC generation by promoting pro-spermatogonia differentiation. Rhox10 also regulates batteries of migration genes and promotes the migration of pro-spermatogonia into the SSC niche. The identification of an X-linked homeobox gene that drives the initial generation of SSCs has implications for the evolution of X-linked gene clusters and sheds light on regulatory mechanisms influencing adult stem cell generation in general.


Subject(s)
Adult Germline Stem Cells/metabolism , Gene Expression Regulation, Developmental , Genes, X-Linked , Homeodomain Proteins/genetics , Spermatogenesis/genetics , Spermatogonia/metabolism , Adult Germline Stem Cells/cytology , Animals , Genes, Developmental , Homeodomain Proteins/metabolism , Male , Mice , Mice, Knockout , Multigene Family , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Spermatogonia/cytology
SELECTION OF CITATIONS
SEARCH DETAIL