[HTML][HTML] The transcription factor Nfatc2 regulates β-cell proliferation and genes associated with type 2 diabetes in mouse and human islets

MP Keller, PK Paul, ME Rabaglia, DS Stapleton… - PLoS …, 2016 - journals.plos.org
MP Keller, PK Paul, ME Rabaglia, DS Stapleton, KL Schueler, AT Broman, SI Ye, N Leng
PLoS genetics, 2016journals.plos.org
Human genome-wide association studies (GWAS) have shown that genetic variation at>
130 gene loci is associated with type 2 diabetes (T2D). We asked if the expression of the
candidate T2D-associated genes within these loci is regulated by a common locus in
pancreatic islets. Using an obese F2 mouse intercross segregating for T2D, we show that
the expression of~ 40% of the T2D-associated genes is linked to a broad region on mouse
chromosome (Chr) 2. As all but 9 of these genes are not physically located on Chr 2, linkage …
Human genome-wide association studies (GWAS) have shown that genetic variation at >130 gene loci is associated with type 2 diabetes (T2D). We asked if the expression of the candidate T2D-associated genes within these loci is regulated by a common locus in pancreatic islets. Using an obese F2 mouse intercross segregating for T2D, we show that the expression of ~40% of the T2D-associated genes is linked to a broad region on mouse chromosome (Chr) 2. As all but 9 of these genes are not physically located on Chr 2, linkage to Chr 2 suggests a genomic factor(s) located on Chr 2 regulates their expression in trans. The transcription factor Nfatc2 is physically located on Chr 2 and its expression demonstrates cis linkage; i.e., its expression maps to itself. When conditioned on the expression of Nfatc2, linkage for the T2D-associated genes was greatly diminished, supporting Nfatc2 as a driver of their expression. Plasma insulin also showed linkage to the same broad region on Chr 2. Overexpression of a constitutively active (ca) form of Nfatc2 induced β-cell proliferation in mouse and human islets, and transcriptionally regulated more than half of the T2D-associated genes. Overexpression of either ca-Nfatc2 or ca-Nfatc1 in mouse islets enhanced insulin secretion, whereas only ca-Nfatc2 was able to promote β-cell proliferation, suggesting distinct molecular pathways mediating insulin secretion vs. β-cell proliferation are regulated by NFAT. Our results suggest that many of the T2D-associated genes are downstream transcriptional targets of NFAT, and may act coordinately in a pathway through which NFAT regulates β-cell proliferation in both mouse and human islets.
PLOS