MAF1 is a conserved nutrient- and stress-sensitive repressor of gene transcription. Best known for its role as a master regulator of RNA polymerase III transcription in yeast, MAF1 has also been shown to affect the expression of select genes transcribed by RNA polymerase II in mammalian cells. MAF1 repression of RNA polymerase III regulates the synthesis of transfer RNAs, 5S rRNA and other abundant non-coding RNAs that together account for ~15% of total RNA in every cell. Thus, MAF1 function is thought to be important for metabolic economy. Recent dramatic proof of this view was provided by the finding that mice with a whole body knockout of Maf1 are lean and profoundly resistant to diet-induced obesity and non-alcoholic fatty liver disease. Maf1 KO mice are metabolically inefficient, have increased energy expenditure and have an extended lifespan indicative of improved health. The current understanding is that increased RNA pol III transcription in every tissue places such a demand on the energetically expensive synthesis of nucleotides that it alters the balance between energy utilization and storage. Thus, the long term goal of this project is to determine how the absence of MAF1 changes gene expression and metabolism to alter metabolic efficiency and energy expenditure.
Aim I of this proposal will examine how gene expression has been altered in key metabolic tissues of Maf1-/- mice by transcriptome and ribosome profiling and by investigating tRNA population effects on translation efficiency and fidelity.
Aim II will determine the metabolic changes underlying the increased energy expenditure of Maf1-/- mice by targeted metabolite profiling. These studies will focus on central metabolic pathways and will be complemented by stable isotope measurements of whole body lipolysis and flux through the pentose phosphate pathway, processes that we hypothesize are associated with the enhanced supply and consumption of metabolic energy in the mice.
Aim III will investigate the role of RNA pol III transcription in driving energy expenditure in Maf1-/- mice through unrestrained synthesis of highly abundant non-coding RNAs, including transfer RNA and non-specific transcription. Nascent elongating transcript sequencing will locate and quantify RNA pol III molecules genome-wide and a new mouse model will be created to suppress Maf1 KO phenotypes that result directly or indirectly from elevated RNA pol III transcription. Finally, the studies in aim IV will determine whether loss of MAF1 in young and adult mice via inducible and systemic Cre- mediated recombination increases metabolic inefficiency. These experiments are expected to support the concept that the postnatal function of MAF1 is critical for metabolic economy while providing a powerful demonstration of the potential of MAF1 as an anti-obesity drug target.

Public Health Relevance

Single gene defects that confer resistance to diet-induced obesity identify potentially important therapeutic targets for the treatment of obesity-related diseases and provide unique opportunities to understand the molecular and metabolic basis of the obesity-resistant phenotype. The focus of this research is a newly identified obesity-resistance gene in the mouse, the transcriptional repressor MAF1. The proposed studies will provide biomedically important knowledge about unique mechanisms for enhancing energy expenditure.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
5R01GM120358-02
Application #
9355692
Study Section
Molecular Genetics A Study Section (MGA)
Program Officer
Sledjeski, Darren D
Project Start
2016-09-21
Project End
2020-07-31
Budget Start
2017-08-01
Budget End
2018-07-31
Support Year
2
Fiscal Year
2017
Total Cost
Indirect Cost
Name
Albert Einstein College of Medicine, Inc
Department
Type
DUNS #
079783367
City
Bronx
State
NY
Country
United States
Zip Code
10461
Willis, Ian M; Moir, Robyn D (2018) Signaling to and from the RNA Polymerase III Transcription and Processing Machinery. Annu Rev Biochem 87:75-100
Willis, Ian M; Moir, Robyn D; Hernandez, Nouria (2018) Metabolic programming a lean phenotype by deregulation of RNA polymerase III. Proc Natl Acad Sci U S A 115:12182-12187
Willis, Ian M (2018) Maf1 phenotypes and cell physiology. Biochim Biophys Acta Gene Regul Mech 1861:330-337
Toledo, Miriam; Batista-Gonzalez, Ana; Merheb, Emilio et al. (2018) Autophagy Regulates the Liver Clock and Glucose Metabolism by Degrading CRY1. Cell Metab 28:268-281.e4
Choquet, Karine; Yang, Sharon; Moir, Robyn D et al. (2017) Absence of neurological abnormalities in mice homozygous for the Polr3a G672E hypomyelinating leukodystrophy mutation. Mol Brain 10:13
Mange, François; Praz, Viviane; Migliavacca, Eugenia et al. (2017) Diurnal regulation of RNA polymerase III transcription is under the control of both the feeding-fasting response and the circadian clock. Genome Res 27:973-984