@article{mousavy_gharavy_sexually_2021, title = {Sexually dimorphic roles for the type 2 diabetes-associated C2cd4b gene in murine glucose homeostasis}, volume = {64}, issn = {0012-186X, 1432-0428}, url = {http://link.springer.com/10.1007/s00125-020-05350-x}, doi = {10.1007/s00125-020-05350-x}, abstract = {Abstract Aims/hypothesis Variants close to the {VPS}13C / C2CD4A/C2CD4B locus are associated with altered risk of type 2 diabetes in genome-wide association studies. While previous functional work has suggested roles for {VPS}13C and C2CD4A in disease development, none has explored the role of C2CD4B . Methods {CRISPR}/Cas9-induced global C2cd4b -knockout mice and zebrafish larvae with c2cd4a deletion were used to study the role of this gene in glucose homeostasis. C2 calcium dependent domain containing protein (C2CD)4A and C2CD4B constructs tagged with {FLAG} or green fluorescent protein were generated to investigate subcellular dynamics using confocal or near-field microscopy and to identify interacting partners by mass spectrometry. Results Systemic inactivation of C2cd4b in mice led to marked, but highly sexually dimorphic changes in body weight and glucose homeostasis. Female C2cd4b mice displayed unchanged body weight compared with control littermates, but abnormal glucose tolerance ({AUC}, p  = 0.01) and defective in vivo, but not in vitro, insulin secretion ( p  = 0.02). This was associated with a marked decrease in follicle-stimulating hormone levels as compared with wild-type ({WT}) littermates ( p  = 0.003). In sharp contrast, male C2cd4b null mice displayed essentially normal glucose tolerance but an increase in body weight ( p  {\textless} 0.001) and fasting blood glucose ( p  = 0.003) after maintenance on a high-fat and -sucrose diet vs {WT} littermates. No metabolic disturbances were observed after global inactivation of C2cd4a in mice, or in pancreatic beta cell function at larval stages in C2cd4a null zebrafish. Fasting blood glucose levels were also unaltered in adult C2cd4a -null fish. C2CD4B and C2CD4A were partially localised to the plasma membrane, with the latter under the control of intracellular Ca 2+ . Binding partners for both included secretory-granule-localised {PTPRN}2/phogrin. Conclusions/interpretation Our studies suggest that C2cd4b may act centrally in the pituitary to influence sex-dependent circuits that control pancreatic beta cell function and glucose tolerance in rodents. However, the absence of sexual dimorphism in the impact of diabetes risk variants argues for additional roles for C2CD4A or {VPS}13C in the control of glucose homeostasis in humans. Data availability The datasets generated and/or analysed during the current study are available in the Biorxiv repository ( www.biorxiv.org/content/10.1101/2020.05.18.099200v1 ). {RNA}-Seq ({GSE}152576) and proteomics ({PXD}021597) data have been deposited to {GEO} ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc={GSE}152576 ) and {ProteomeXchange} ( www.ebi.ac.uk/pride/archive/projects/{PXD}021597 ) repositories, respectively. Graphical abstract}, pages = {850--864}, number = {4}, journaltitle = {Diabetologia}, shortjournal = {Diabetologia}, author = {Mousavy Gharavy, S. Neda and Owen, Bryn M. and Millership, Steven J. and Chabosseau, Pauline and Pizza, Grazia and Martinez-Sanchez, Aida and Tasoez, Emirhan and Georgiadou, Eleni and Hu, Ming and Fine, Nicholas H. F. and Jacobson, David A. and Dickerson, Matthew T. and Idevall-Hagren, Olof and Montoya, Alex and Kramer, Holger and Mehta, Zenobia and Withers, Dominic J. and Ninov, Nikolay and Gadue, Paul J. and Cardenas-Diaz, Fabian L. and Cruciani-Guglielmacci, Céline and Magnan, Christophe and Ibberson, Mark and Leclerc, Isabelle and Voz, Marianne and Rutter, Guy A.}, urldate = {2021-12-07}, date = {2021-04}, langid = {english}, keywords = {{WP}4}, }