Sperm RNA code programmes the metabolic health of offspring

Yunfang Zhang, Junchao Shi, Minoo Rassoulzadegan, Francesca Tuorto, Qi Chen, Yunfang Zhang, Junchao Shi, Minoo Rassoulzadegan, Francesca Tuorto, Qi Chen

Abstract

Mammalian sperm RNA is increasingly recognized as an additional source of paternal hereditary information beyond DNA. Environmental inputs, including an unhealthy diet, mental stresses and toxin exposure, can reshape the sperm RNA signature and induce offspring phenotypes that relate to paternal environmental stressors. Our understanding of the categories of sperm RNAs (such as tRNA-derived small RNAs, microRNAs, ribosomal RNA-derived small RNAs and long non-coding RNAs) and associated RNA modifications is expanding and has begun to reveal the functional diversity and information capacity of these molecules. However, the coding mechanism endowed by sperm RNA structures and by RNA interactions with DNA and other epigenetic factors remains unknown. How sperm RNA-encoded information is decoded in early embryos to control offspring phenotypes also remains unclear. Complete deciphering of the 'sperm RNA code' with regard to metabolic control could move the field towards translational applications and precision medicine, and this may lead to prevention of intergenerational transmission of obesity and type 2 diabetes mellitus susceptibility.

Conflict of interest statement

Competing interests

The authors declare no competing interests.

Figures

Fig. 1 |. Information capacity and functional…
Fig. 1 |. Information capacity and functional specificity of the sperm RNA code.
Schematic representation of the sperm RNA code (an interconnected combination of RNA expression and RNA modification profile) that reflects paternal environmental exposure and mediates the transmission of specific phenotypes to the offspring. Different paternal information can be encoded in specific subsets of sperm RNA fractions. lncRNA, long non-coding RNA; miRNA, microRNA; piRNA, Piwi-interacting RNA; rsRNA, ribosomal RNA-derived small RNA; tsRNA, tRNA-derived small RNA.
Fig. 2 |. Potential mechanisms in transformation…
Fig. 2 |. Potential mechanisms in transformation of the sperm RNA code during embryo development.
a | In mouse early embryo, the tRNA-derived small RNA (tsRNA) expression level increases from the four-cell to eight-cell transition, and the tsRNA composition is similar to that of mature sperm. At certain genomic loci, the H3K4me3 pattern in sperm is initially removed in zygotes but re-established in both paternal and maternal chromosomes from the late two-cell embryo stage,. It would be interesting to explore the causal relationship between tsRNAs and the H3K4me3 pattern in sperm and embryo. b | The hypothesis of tsRNA-mediated ribosome heterogeneity that generates biased translational specificity towards different pools of mRNA subpopulations. c | A hypothetical scenario to explain how the initial abnormal metabolic transcriptome induced by sperm RNA can be maintained throughout embryo development. This could be triggered by a self-amplifying loop of abnormal metabolic transcriptome, metabolites and epigenome during embryo development. Acyl-CoA, acetyl coenzyme A; HFD, high-fat diet; SAM, S-adenosyl methionine.
Fig. 3 |. Future applications and precision…
Fig. 3 |. Future applications and precision medicine based on high resolution of sperm RNA code.
a | Future application of third-generation RNA-sequencing platform to simultaneously obtain full RNA sequences and meanwhile pinpoint different RNA modifications at base resolution. b | Future use of sperm RNA code information as clinical guidance for well-timed pregnancy and to reduce the susceptibility to metabolic disorder in the offspring.

References

    1. Barres R & Zierath JR The role of diet and exercise in the transgenerational epigenetic landscape of T2DM. Nat. Rev. Endocrinol 12, 441–451 (2016).
    1. Sales VM, Ferguson-Smith AC & Patti ME Epigenetic mechanisms of transmission of metabolic disease across generations. Cell Metab 25, 559–571 (2017).
    1. Fleming TP et al. Origins of lifetime health around the time of conception: causes and consequences. Lancet 391, 1842–1852 (2018).
    1. Eichler EE et al. Missing heritability and strategies for finding the underlying causes of complex disease. Nat. Rev. Genet 11, 446–450 (2010).
    1. Skvortsova K, Iovino N & Bogdanovic O Functions and mechanisms of epigenetic inheritance in animals. Nat. Rev. Mol. Cell Biol 19, 774–790 (2018).
    1. Miska EA & Ferguson-Smith AC Transgenerational inheritance: models and mechanisms of non-DNA sequence-based inheritance. Science 354, 59–63 (2016).
    1. Heard E & Martienssen RA Transgenerational epigenetic inheritance: myths and mechanisms. Cell 157, 95–109 (2014).
    1. Chen Q, Yan W & Duan E Epigenetic inheritance of acquired traits through sperm RNAs and sperm RNA modifications. Nat. Rev. Genet 17, 733–743 (2016).
    1. Perez MF & Lehner B Intergenerational and transgenerational epigenetic inheritance in animals. Nat. Cell Biol 21, 143–151 (2019).
    1. Bouret S, Levin BE & Ozanne SE Gene-environment interactions controlling energy and glucose homeostasis and the developmental origins of obesity. Physiol. Rev 95, 47–82 (2015).
    1. Oestreich AK & Moley KH Developmental and transmittable origins of obesity-associated health disorders. Trends Genet 33, 399–407 (2017).
    1. Godfrey KM et al. Influence of maternal obesity on the long-term health of offspring. Lancet Diabetes Endocrinol. 5, 53–64 (2017).
    1. Radford EJ et al. In utero effects. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 345, 1255903 (2014).
    1. Kazachenka A et al. Identification, characterization, and heritability of murine metastable epialleles: implications for non-genetic inheritance. Cell 175, 1259–1271 (2018).
    1. Hammoud SS et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).
    1. Brykczynska U et al. Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol 17, 679–687 (2010).
    1. Siklenka K et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 350, aab2006 (2015).
    1. Chen Q et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science 351, 397–400 (2016).
    1. Gapp K et al. Alterations in sperm long RNA contribute to the epigenetic inheritance of the effects of postnatal trauma. Mol. Psychiatry 10.1038/s41380-018-0271-6 (2018).
    1. Grandjean V et al. RNA-mediated paternal heredity of diet-induced obesity and metabolic disorders. Sci. Rep 5, 18193 (2015).
    1. Gapp K et al. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat. Neurosci 17, 667–669 (2014).
    1. Zhang Y et al. Dnmt2 mediates intergenerational transmission of paternally acquired metabolic disorders through sperm small non-coding RNAs. Nat. Cell Biol 20, 535–540 (2018).
    1. Johnson GD et al. Chromatin and extracellular vesicle associated sperm RNAs. Nucleic Acids Res 43, 6847–6859 (2015).
    1. Peng H et al. A novel class of tRNA-derived small RNAs extremely enriched in mature mouse sperm. Cell Res 22, 1609–1612 (2012).
    1. Sharma U et al. Small RNAs are trafficked from the epididymis to developing mammalian sperm. Dev. Cell 46, 481–494 (2018).
    1. Sharma U et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 351, 391–396 (2016).
    1. Chu C et al. A sequence of 28S rRNA-derived small RNAs is enriched in mature sperm and various somatic tissues and possibly associates with inflammation. J. Mol. Cell Biol 9, 256–259 (2017).
    1. Shi J et al. SPORTS1.0: a tool for annotating and profiling non-coding RNAs optimized for rRNA-and tRNA-derived small RNAs. Genomics Proteomics Bioinformatics 16, 144–151 (2018).
    1. Schuster A et al. SpermBase: a database for sperm-borne RNA contents. Biol. Reprod 95, 99 (2016).
    1. Hua M et al. Identification of small non-coding RNAs as sperm quality biomarkers for in vitro fertilization. Cell Discov 5, 20 (2019).
    1. Schimmel P The emerging complexity of the tRNA world: mammalian tRNAs beyond protein synthesis. Nat. Rev. Mol. Cell Biol 19, 45–58 (2018).
    1. Guo L et al. Sperm-carried RNAs play critical roles in mouse embryonic development. Oncotarget 8, 67394–67405 (2017).
    1. Yuan S et al. Sperm-borne miRNAs and endo-siRNAs are important for fertilization and preimplantation embryonic development. Development 143, 635–647 (2016).
    1. Frye M et al. RNA modifications modulate gene expression during development. Science 361, 1346–1349 (2018).
    1. Pan T Modifications and functional genomics of human transfer RNA. Cell Res 28, 395–404 (2018).
    1. Guzzi N et al. Pseudouridylation of tRNA-derived fragments steers translational control in stem cells.Cell 173, 1204–1216 (2018).
    1. Safra M et al. The m1A landscape on cytosolic and mitochondrial mRNA at single-base resolution. Nature 551, 251–255 (2017).
    1. Murashov AK et al. Paternal long-term exercise programs offspring for low energy expenditure and increased risk for obesity in mice. FASEB J 30, 775–784 (2016).
    1. Rompala GR et al. Heavy chronic intermittent ethanol exposure alters small noncoding RNAs in mouse sperm and epididymosomes. Front. Genet 9, 32 (2018).
    1. Rassoulzadegan M et al. RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse. Nature 441, 469–474 (2006).
    1. Kiani J et al. RNA-mediated epigenetic heredity requires the cytosine methyltransferase Dnmt2. PLoS Genet 9, e1003498 (2013).
    1. Schulz NKE, Diddens-de Buhr MF & Kurtz J Paternal knockdown of Dnmt2 increases offspring susceptibility to bacterial infection. Preprint at bioRxiv (2018).
    1. Lyko F The DNA methyltransferase family: a versatile toolkit for epigenetic regulation. Nat. Rev. Genet 19, 81–92 (2018).
    1. Zhang X et al. Small RNA modifications: integral to function and disease. Trends Mol. Med 22, 1025–1034 (2016).
    1. Schaefer M et al. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev 24, 1590–1595 (2010).
    1. Tuorto F et al. RNA cytosine methylation by Dnmt2 and NSun2 promotes tRNA stability and protein synthesis. Nat. Struct. Mol. Biol 19, 900–905 (2012).
    1. Legrand C et al. Statistically robust methylation calling for whole-transcriptome bisulfite sequencing reveals distinct methylation patterns for mouse RNAs. Genome Res 27, 1589–1596 (2017).
    1. Tuorto F et al. Queuosine-modified tRNAs confer nutritional control of protein translation. EMBO J 37, e99777 (2018).
    1. Muller M et al. Dynamic modulation of Dnmt2-dependent tRNA methylation by the micronutrient queuine. Nucleic Acids Res 43, 10952–10962 (2015).
    1. Wang X et al. Queuosine modification protects cognate tRNAs against ribonuclease cleavage. RNA 24, 1305–1313 (2018).
    1. Shi J et al. tsRNAs: the Swiss army knife for translational regulation. Trends Biochem. Sci 44, 185–189 (2018).
    1. Wang X et al. Transcriptome-wide reprogramming of N(6)-methyladenosine modification by the mouse microbiome. Cell Res 29, 167–170 (2019).
    1. Nelson VR et al. Transgenerational epigenetic 73. effects of the Apobec1 cytidine deaminase deficiency on testicular germ cell tumor susceptibility and embryonic viability. Proc. Natl Acad. Sci. U. S. A 109, 74 E2766–E2773 (2012).
    1. Liu WM et al. Sperm-borne microRNA-34c is required for the first cleavage division in mouse. Proc. Natl Acad. 75. Sci. U. S. A 109, 490–494 (2012).
    1. Yuan S et al. mir-34b/c and mir-449a/b/c are required for spermatogenesis, but not for the first cleavage division in mice. Biol. Open 4, 212–223 (2015). 76.
    1. Conine CC et al. Small RNAs gained during epididymal transit of sperm are essential for embryonic development in mice. Dev. Cell 46, 470–480 (2018).
    1. Suganuma R, Yanagimachi R & Meistrich ML 77. Decline in fertility of mouse sperm with abnormal chromatin during epididymal passage as revealed by ICSI. Hum. Reprod 20, 3101–3108 (2005). 78.
    1. Cheng HS et al. Increased susceptibility of post-weaning rats on high-fat diet to metabolic syndrome. J. Adv. Res 8, 743–752 (2017). 79.
    1. Sarker G et al. Maternal overnutrition programs hedonic and metabolic phenotypes across generations through sperm tsRNAs. Proc. Natl Acad. Sci. U. S. A 80116, 10547–10556 (2019).
    1. Rechavi O & Lev I Principles of transgenerational small RNA inheritance in Caenorhabditis elegans. Curr. Biol 27, R720–R730 (2017). 81.
    1. Yu R, Wang X & Moazed D Epigenetic inheritance mediated by coupling of RNAi and histone H3K9 methylation. Nature 558, 615–619 (2018).
    1. Motamedi MR et al. Two RNAi complexes, RITS and RDRC, physically interact and localize to noncoding 82. centromeric RNAs. Cell 119, 789–802 (2004).
    1. Ost A et al. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell 159, 1352–1364 (2014).
    1. Daxinger L et al. Hypomethylation of ERVs in the 83. sperm of mice haploinsufficient for the histone methyltransferase Setdb1 correlates with a paternal effect on phenotype. Sci. Rep 6, 25004 (2016). 84.
    1. Morgan HD et al. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet 23, 314–318 (1999). 85.
    1. Schorn AJ et al. LTR-retrotransposon control by tRNA-derived small RNAs. Cell 170, 61–71 (2017).
    1. Genenncher B et al. Mutations in cytosine-5 tRNA methyltransferases impact mobile element expression 86. and genome stability at specific DNA repeats. Cell Rep 22, 1861–1874 (2018).
    1. Martinez G tRNA-derived small RNAs: new players 87. in genome protection against retrotransposons. RNA Biol 15, 170–175 (2018).
    1. Zhang Y, Shi J & Chen Q tsRNAs: new players in 88. mammalian retrotransposon control. Cell Res 27, 1307–1308 (2017).
    1. Schorn AJ & Martienssen R Tie-break: host and 89. retrotransposons play tRNA. Trends Cell Biol 28, 793–806 (2018). 90.
    1. Horie M et al. An RNA-dependent RNA polymerase gene in bat genomes derived from an ancient negative-strand RNA virus. Sci. Rep 6, 25873 (2016). 91.
    1. Yang Q et al. Highly sensitive sequencing reveals dynamic modifications and activities of small RNAs in mouse oocytes and early embryos. Sci. Adv 2, 92 e1501482 (2016).
    1. Zhang B et al. Allelic reprogramming of the histone modification H3K4me3 in early mammalian development. Nature 537, 553–557 (2016).
    1. Dahl JA et al. Broad histone H3K4me3 domains in mouse oocytes modulate maternal-to-zygotic transition. Nature 537, 548–552 (2016).
    1. van de Werken C et al. Paternal heterochromatin formation in human embryos is H3K9/HP1 directed and primed by sperm-derived histone modifications. Nat. Commun 5, 5868 (2014).
    1. Genuth NR & Barna M The discovery of ribosome heterogeneity and its implications for gene regulation and organismal life. Mol. Cell 71, 364–374 (2018).
    1. Shi Z et al. Heterogeneous ribosomes preferentially translate distinct subpools of mRNAs genome-wide. Mol. Cell 67, 71–83 (2017).
    1. Li X et al. Regulation of chromatin and gene expression by metabolic enzymes and metabolites. Nat. Rev. Mol. Cell Biol 19, 563–578 (2018).
    1. Chen Q et al. Tracing the origin of heterogeneity and symmetry breaking in the early mammalian embryo. Nat. Commun 9, 1819 (2018).
    1. Shi J et al. Dynamic transcriptional symmetry-breaking in pre-implantation mammalian embryo development revealed by single-cell RNA-seq. Development 142, 3468–3477 (2015).
    1. Skinner MK et al. Alterations in sperm DNA methylation, non-coding RNA and histone retention associate with DDT-induced epigenetic transgenerational inheritance of disease. Epigenetics Chromatin 11, 8 (2018).
    1. Schuster A, Skinner MK & Yan W Ancestral vinclozolin exposure alters the epigenetic transgenerational inheritance of sperm small noncoding RNAs. Environ. Epigenet 2, dvw001 (2016).
    1. Donkin I et al. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in humans. CellMetab 23, 369–378 (2016).
    1. Stanford KI et al. Paternal exercise improves glucose metabolism in adult offspring. Diabetes 67, 2530–2540 (2018).
    1. Ingerslev LR et al. Endurance training remodels sperm-borne small RNA expression and methylation at neurological gene hotspots. Clin. Epigenet 10, 12 (2018).
    1. Kwok CK et al. rG4-seq reveals widespread formation of G-quadruplex structures in the human transcriptome. Nat. Methods 13, 841–844 (2016).
    1. Goodwin S, McPherson JD & McCombie WR Coming of age: ten years of next-generation sequencing technologies. Nat. Rev. Genet 17, 333–351 (2016).
    1. Zhang Y & Chen Q The expanding repertoire of hereditary information carriers. Development 146, dev170902 (2019).
    1. Dekker J et al. The 4D nucleome project. Nature 549, 219–226 (2017).
    1. Guo F et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res 27, 967–988 (2017).
    1. Bian S et al. Single-cell multiomics sequencing and analyses of human colorectal cancer. Science 362, 1060–1063 (2018).
    1. Li X et al. GRID-seq reveals the global RNA-chromatin interactome. Nat. Biotechnol 35, 940–950 (2017).
    1. Lu Z et al. RNA duplex map in living cells reveals higher-order transcriptome structure. Cell 165, 1267–1279 (2016).
    1. Topol EJ High-performance medicine: the convergence of human and artificial intelligence. Nat. Med 25, 44–56 (2019).
    1. Rodriguez KF et al. Effects of in utero exposure to arsenic during the second half of gestation on reproductive end points and metabolic parameters in female CD-1 mice. Environ. Health Perspect 124, 336–343 (2016).
    1. Dias BG & Ressler KJ Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat. Neurosci 17, 89–96 (2014).
    1. Sun W et al. Cold-induced epigenetic programming of the sperm enhances brown adipose tissue activity in the offspring. Nat. Med 24, 1372–1383 (2018).
    1. Le Q et al. Drug-seeking motivation level in male rats determines offspring susceptibility or resistance to cocaine-seeking behaviour. Nat. Commun 8, 15527 (2017).
    1. Yan W et al. Birth of mice after intracytoplasmic injection of single purified sperm nuclei and detection of messenger RNAs and MicroRNAs in the sperm nuclei. Biol. Reprod 78, 896–902 (2008).
    1. Lalancette C et al. Paternal contributions: new functional insights for spermatozoal RNA. J. Cell. Biochem 104, 1570–1579 (2008).
    1. Hamatani T Human spermatozoal RNAs. Fertil. Steril 97, 275–281 (2012).
    1. Al-Dossary AA et al. Oviductosome-sperm membrane interaction in cargo delivery: detection of fusion and underlying molecular players using three-dimensional super-resolution structured illumination microscopy (SR-SIM). J. Biol. Chem 290, 17710–17723 (2015).
    1. Liu Y & Chen Q 150 years of Darwin’s theory of intercellular flow of hereditary information. Nat. Rev. Mol. Cell Biol 19, 749–750 (2018).

Source: PubMed

3
Abonnere