Alternative splicing of helicase-like transcription factor (Hltf): Intron retention-dependent activation of immune tolerance at the feto-maternal interface

PLoS One. 2018 Jul 5;13(7):e0200211. doi: 10.1371/journal.pone.0200211. eCollection 2018.

Abstract

Hltf is regulated by intron retention, and global Hltf-deletion causes perinatal lethality from hypoglycemia. In heart, full-length Hltf is a transcriptional regulator of Hif-1α that controls transport systems. Thus, we tested the hypothesis that Hltf deletion from placenta caused or exacerbated neonatal hypoglycemia via Hif-1α regulation of nutrient transporters. RNA-seq data analyses identified significant changes in transcript expression and alternative splicing (AS) in E18.5 placentome. iPathwayGuide was used for gene ontology (GO) analysis of biological processes, molecular functions and cellular components. Elim pruning algorithm identified hierarchical relationships. The methylome was interrogated by Methyl-MiniSeq Epiquest analysis. GO analysis identified gene enrichment within biological processes. Protein expression was visualized with immunohistochemistry. Although two Hltf mRNA isoforms are quantifiable in most murine tissues, only the truncated Hltf isoform is expressed in placenta. The responsible intron retention event occurs in the absence of DNA methylation. iPathwayGuide analysis identified 157 target genes of 11,538 total genes with measured expression. These were obtained using a threshold of 0.05 for statistical significance (p-value) and a long fold change of expression with absolute value of at least 0.6. Hltf deletion altered transcription of trophoblast lineage-specific genes, and increased transcription of the Cxcr7 (p = 0.004) gene whose protein product is a co-receptor for human and simian immunodeficiency viruses. Concomitant increased Cxcr7 protein was identified with immunolabeling. Hltf deletion had no effect on transcription or site-specific methylation patterns of Hif-1α, the major glucose transporters, or System A amino acid transporters. There was no measureable evidence of uteroplacental dysfunction or fetal compromise. iPathGuide analysis revealed Hltf suppresses cytolysis (10/21 genes; p-value 1.900e-12; p-value correction: Elim pruning; GO:019835) including the perforin-granzyme pathway in uterine natural killer cells. Our findings 1) prove the truncated Hltf protein isoform is a transcription factor, 2) establish a functional link between AS of Hltf and immunosuppression at the feto-maternal interface, 3) correlate intron retention with the absence of DNA methylation, and 4) underscore the importance of differential splicing analysis to identify Hltf's functional diversity.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Alternative Splicing
  • Animals
  • Carrier Proteins
  • DNA Methylation
  • DNA-Binding Proteins / genetics*
  • DNA-Binding Proteins / immunology*
  • Exons
  • Female
  • Fetomaternal Transfusion / genetics
  • Fetomaternal Transfusion / pathology
  • Gene Expression Profiling
  • Immune Tolerance / genetics*
  • Introns
  • Maternal-Fetal Exchange / immunology*
  • Mice
  • Mice, Inbred C57BL
  • Placenta / immunology*
  • Pregnancy
  • Protein Isoforms
  • Receptors, CXCR / genetics
  • Receptors, CXCR / immunology
  • Transcription Factors / genetics*
  • Transcription Factors / immunology*

Substances

  • Carrier Proteins
  • Cmkor1 protein, mouse
  • DNA-Binding Proteins
  • Protein Isoforms
  • Receptors, CXCR
  • Smarca3 protein, mouse
  • Transcription Factors

Grants and funding

Funding sources were all local or institutional and included the South Plains Foundation, TTUHSC collaborative research grant from the School of Medicine, and the Laura W. Bush Institute for Women's Health research program from the University Medical Center. The funders had no role in study design, data collection, and analysis, decision to publish, or preparation of the manuscript.