@article {Grzyb2021.06.04.447058, author = {Anna N. Grzyb and Rupert W. Overall and Jan Silhavy and Petr Mlejnek and Hana Mal{\'\i}nsk{\'a} and Martina H{\"u}ttl and Irena Markov{\'a} and Klaus S. Fabel and Lu Lu and Ales Stuchlik and Robert W. Williams and Michal Pravenec and Gerd Kempermann}, title = {System genetics in the rat HXB/BXH family identifies Tti2 as a pleiotropic quantitative trait gene for adult hippocampal neurogenesis and serum glucose}, elocation-id = {2021.06.04.447058}, year = {2021}, doi = {10.1101/2021.06.04.447058}, publisher = {Cold Spring Harbor Laboratory}, abstract = {Neurogenesis in the adult hippocampus contributes to learning and memory in the healthy brain but is dysregulated in metabolic and neurodegenerative diseases. The molecular relationships between neural stem cell activity, adult neurogenesis, and global metabolism are largely unknown. Here we applied unbiased systems genetic methods to quantify genetic covariation among adult neurogenesis and metabolic phenotypes in peripheral tissues of a genetically diverse family of rat strains, derived from a cross between the spontaneously hypertensive (SHR/OlaIpcv) strain and Brown Norway (BN-Lx/Cub). The HXB/BXH family is a very well established model to dissect genetic variants that modulate metabolic and cardiovascular disease and we have accumulated deep phenome and transcriptome data in a FAIR-compliant resource for systematic and integrative analyses. Here we measured rates of precursor cell proliferation, survival of new neurons, and gene expression in the hippocampus of the entire HXB/BXH family, including both parents. These data were combined with published metabolic phenotypes to detect a neurometabolic quantitative trait locus (QTL) for serum glucose and neuronal survival. We subsequently fine-mapped a key phenotype to a locus that includes the telo2-interacting protein 2 gene (Tti2){\textemdash}a chaperone that modulates the activity and stability of PIKK kinases. To validate variants in or near Tti2 as a cause for differences in neurogenesis and glucose levels, we generated a targeted frameshift mutation on the SHR/OlaIpcv background. Heterozygous SHR-Tti2+/- mutants had lower rates of hippocampal neurogenesis and hallmarks of dysglycemia compared to wild-type littermates. Our findings highlight Tti2 as a causal genetic and molecular link between glucose metabolism and structural brain plasticity. In humans, more than 800 genomic variants are linked to TTI2 expression, seven of which have associations to protein and blood stem cell factor concentrations, blood pressure and frontotemporal dementia.Author summary Metabolic and neurological disorders are often comorbid, suggesting that biological pathways which orchestrate peripheral homeostasis and the integrity of the nervous system intersect. The genetic architecture behind these relationships is still poorly described, in part because molecular processes in the human brain are very difficult to study. We thus used a rodent genetic reference population to investigate links between adult hippocampal neurogenesis{\textemdash} a cellular plasticity mechanism important for learning flexibility{\textemdash}and metabolism. We measured adult neurogenesis in the family of 30 HXB/BXH rat recombinant inbred strains, who are characterised by stable differences in metabolism, behaviour, and gene expression levels. Because gene variants affecting distinct traits segregated into different members of the family, we discovered that previously published phenotypes correlated to adult neurogenesis due to shared genomic sequence. We found that expression levels of Tti2{\textemdash}a part of a specialised protein chaperone complex regulating stability of PIKK kinases{\textemdash}were concomitantly influencing adult neurogenesis and serum glucose levels. In human populations hundreds of genomic variants regulate TTI2 expression, potentially affecting brain function and glucose homeostasis.Competing Interest StatementThe authors have declared no competing interest.}, URL = {https://www.biorxiv.org/content/early/2021/06/04/2021.06.04.447058}, eprint = {https://www.biorxiv.org/content/early/2021/06/04/2021.06.04.447058.full.pdf}, journal = {bioRxiv} }