Abstract

Single-nucleotide polymorphisms within intron 1 of the FTO (fat mass and obesity-associated) gene are associated with enhanced FTO expression, increased body weight, obesity and type 2 diabetes mellitus (T2DM). The N6-methyladenosine (m6A) demethylase FTO plays a pivotal regulatory role for postnatal growth and energy expenditure. The purpose of this review is to provide translational evidence that links milk signaling with FTO-activated transcription of the milk recipient. FTO-dependent demethylation of m6A regulates mRNA splicing required for adipogenesis, increases the stability of mRNAs, and affects microRNA (miRNA) expression and miRNA biosynthesis. FTO senses branched-chain amino acids (BCAAs) and activates the nutrient sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1), which plays a key role in translation. Milk provides abundant BCAAs and glutamine, critical components increasing FTO expression. CpG hypomethylation in the first intron of FTO has recently been associated with T2DM. CpG methylation is generally associated with gene silencing. In contrast, CpG demethylation generally increases transcription. DNA de novo methylation of CpG sites is facilitated by DNA methyltransferases (DNMT) 3A and 3B, whereas DNA maintenance methylation is controlled by DNMT1. MiRNA-29s target all DNMTs and thus reduce DNA CpG methylation. Cow´s milk provides substantial amounts of exosomal miRNA-29s that reach the systemic circulation and target mRNAs of the milk recipient. Via DNMT suppression, milk exosomal miRNA-29s may reduce the magnitude of FTO methylation, thereby epigenetically increasing FTO expression in the milk consumer. High lactation performance with increased milk yield has recently been associated with excessive miRNA-29 expression of dairy cow mammary epithelial cells (DCMECs). Notably, the galactopoietic hormone prolactin upregulates the transcription factor STAT3, which induces miRNA-29 expression. In a retrovirus-like manner milk exosomes may transfer DCMEC-derived miRNA-29s and bovine FTO mRNA to the milk consumer amplifying FTO expression. There is compelling evidence that obesity, T2DM, prostate and breast cancer, and neurodegenerative diseases are all associated with increased FTO expression. Maximization of lactation performance by veterinary medicine with enhanced miRNA-29s and FTO expression associated with increased exosomal miRNA-29 and FTO mRNA transfer to the milk consumer may represent key epigenetic mechanisms promoting FTO/mTORC1-mediated diseases of civilization.

Highlights

  • FTO, fat mass- and obesity-associated gene (MIM 612938) maps to chromosome 16q12.2 and is widely expressed in a variety of human tissues with highestMelnik J Transl Med (2015) 13:385 first intron of FTO are highly associated with obesity and obesity-related traits [1, 6, 7]

  • Current research focuses on the role of epigenetic causes of excess adult weight gain and early onset of age-related diseases [337]

  • The FTO gene has been recognized to play a crucial role in the early-life determination of body weight, body composition and energy balance [337]

Read more

Summary

Introduction

FTO, fat mass- and obesity-associated gene (MIM 612938) maps to chromosome 16q12.2 and is widely expressed in a variety of human tissues with highestMelnik J Transl Med (2015) 13:385 first intron of FTO are highly associated with obesity and obesity-related traits [1, 6, 7]. FTO, fat mass- and obesity-associated gene (MIM 612938) maps to chromosome 16q12.2 and is widely expressed in a variety of human tissues with highest. Obesity is a well-known risk factor for the development of type 2 diabetes mellitus (T2DM). Genetic polymorphisms of FTO, and the methylation status of FTO, especially CpG hypomethylation of intron 1 has been linked to increased T2DM prevalence [31]. It is not known whether demethylated CpG loci in intron 1 map directly to regulatory regions and SNPs. Notably, FTO methylation in human pancreatic islets of T2DM patients is significantly reduced compared to healthy controls [32]. It is well appreciated that dietary factors induce epigenetic alterations, which have pivotal longterm biological consequences [33]

Objectives
Findings
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