Expression and functional assessment of candidate type 2 diabetes susceptibility genes identify four new genes contributing to human insulin secretion

Fatou K Ndiaye, Ana Ortalli, Mickaël Canouil, Marlène Huyvaert, Clara Salazar-Cardozo, Cécile Lecoeur, Marie Verbanck, Valérie Pawlowski, Raphaël Boutry, Emmanuelle Durand, Iandry Rabearivelo, Olivier Sand, Lorella Marselli, Julie Kerr-Conte, Vikash Chandra, Raphaël Scharfmann, Odile Poulain-Godefroy, Piero Marchetti, François Pattou, Amar Abderrahmani, Philippe Froguel, Amélie Bonnefond, Fatou K Ndiaye, Ana Ortalli, Mickaël Canouil, Marlène Huyvaert, Clara Salazar-Cardozo, Cécile Lecoeur, Marie Verbanck, Valérie Pawlowski, Raphaël Boutry, Emmanuelle Durand, Iandry Rabearivelo, Olivier Sand, Lorella Marselli, Julie Kerr-Conte, Vikash Chandra, Raphaël Scharfmann, Odile Poulain-Godefroy, Piero Marchetti, François Pattou, Amar Abderrahmani, Philippe Froguel, Amélie Bonnefond

Abstract

Objectives: Genome-wide association studies (GWAS) have identified >100 loci independently contributing to type 2 diabetes (T2D) risk. However, translational implications for precision medicine and for the development of novel treatments have been disappointing, due to poor knowledge of how these loci impact T2D pathophysiology. Here, we aimed to measure the expression of genes located nearby T2D associated signals and to assess their effect on insulin secretion from pancreatic beta cells.

Methods: The expression of 104 candidate T2D susceptibility genes was measured in a human multi-tissue panel, through PCR-free expression assay. The effects of the knockdown of beta-cell enriched genes were next investigated on insulin secretion from the human EndoC-βH1 beta-cell line. Finally, we performed RNA-sequencing (RNA-seq) so as to assess the pathways affected by the knockdown of the new genes impacting insulin secretion from EndoC-βH1, and we analyzed the expression of the new genes in mouse models with altered pancreatic beta-cell function.

Results: We found that the candidate T2D susceptibility genes' expression is significantly enriched in pancreatic beta cells obtained by laser capture microdissection or sorted by flow cytometry and in EndoC-βH1 cells, but not in insulin sensitive tissues. Furthermore, the knockdown of seven T2D-susceptibility genes (CDKN2A, GCK, HNF4A, KCNK16, SLC30A8, TBC1D4, and TCF19) with already known expression and/or function in beta cells changed insulin secretion, supporting our functional approach. We showed first evidence for a role in insulin secretion of four candidate T2D-susceptibility genes (PRC1, SRR, ZFAND3, and ZFAND6) with no previous knowledge of presence and function in beta cells. RNA-seq in EndoC-βH1 cells with decreased expression of PRC1, SRR, ZFAND6, or ZFAND3 identified specific gene networks related to T2D pathophysiology. Finally, a positive correlation between the expression of Ins2 and the expression of Prc1, Srr, Zfand6, and Zfand3 was found in mouse pancreatic islets with altered beta-cell function.

Conclusions: This study showed the ability of post-GWAS functional studies to identify new genes and pathways involved in human pancreatic beta-cell function and in T2D pathophysiology.

Keywords: EndoC-βH1; Expression analysis; Genome-wide association study; Insulin secretion; RNAi screening; Type 2 diabetes.

Figures

Figure 1
Figure 1
Enrichment analysis of the expression of the genes causing monogenic diabetes in the panel of human organs, tissues, and cells. The black vertical line denotes a p-value of 0.05.
Figure 2
Figure 2
Enrichment analysis of the expression of candidate T2D-susceptibility genes in the panel of human organs, tissues, and cells. The black vertical line denotes a p-value of 0.05.
Figure 3
Figure 3
Decreased expression of HNF4A (A) or GCK (B), causing monogenic diabetes, leads to impaired insulin secretion from EndoC-βH1 cells. EndoC-βH1 cells were transfected with control non-targeting pool siRNA (siNTP) or target gene siRNA and were analyzed 72 h post-transfection. Insulin secretion (percentage of secretion of the total insulin content) was analyzed in response to 60 min incubation with 0.5 mM glucose (±0.5 mM IBMX), followed by 60 min incubation with 16.7 mM glucose (±0.5 mM IBMX). Data represent mean values ± SEM of at least three independent experiments. **p < 0.01; ***p < 0.001; ns, not significant. Glc, glucose; IBMX, phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine.
Figure 4
Figure 4
Decreased expression of TCF19 (A), SLC30A8 (B), TBC1D4 (C), CDKN2A (D), or KCNK16 (E), already suggested to be expressed in beta cell and/or to contribute to beta-cell function, significantly modifies insulin secretion from EndoC-βH1 cells. EndoC-βH1 cells were transfected with control non-targeting pool siRNA (siNTP) or target gene siRNA and were analyzed 72 h post-transfection. Insulin secretion (percentage of secretion of the total insulin content) was analyzed in response to 60 min incubation with 0.5 mM glucose (±0.5 mM IBMX), followed by 60 min incubation with 16.7 mM glucose (±0.5 mM IBMX). Data represent mean values ± SEM of at least three independent experiments. *p < 0.05; **p < 0.01; ns, not significant. Glc, glucose; IBMX, phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine.
Figure 5
Figure 5
Decreased expression of PRC1 (A), SRR (B), ZFAND3 (C), or ZFAND6 (D), with no evidence of expression and role in pancreatic beta cells, leads to impaired insulin secretion from EndoC-βH1 cells. EndoC-βH1 cells were transfected with control non-targeting pool siRNA (siNTP) or target gene siRNA and were analyzed 72 h post-transfection. Insulin secretion (percentage of secretion of the total insulin content) was analyzed in response to 60 min incubation with 0.5 mM glucose (±0.5 mM IBMX), followed by 60 min incubation with 16.7 mM glucose (±0.5 mM IBMX). Data represent mean values ± SEM of at least three independent experiments. *p < 0.05; **p < 0.01; ns, not significant. Glc, glucose; IBMX, phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine.
Figure 6
Figure 6
PRC1 (A), SRR (B), ZFAND3 (C), and ZFAND6 (D) are expressed in human pancreatic islets and beta cells. Representative images of immunofluorescence staining for PRC1 (green; A), SRR (green; B), ZFAND3 (green; C), ZFAND6 (green, D), and insulin (red) performed on fixed isolated human islet clusters. Yellow (merged images) indicates co-localization of PRC1, SRR, ZFAND3, or ZFAND6 with insulin. Blue, DAPI.
Figure 7
Figure 7
Decreased PRC1 expression in EndoC-βH1 cells leads to increased apoptosis and decreased cell viability. List of deregulated genes emphasized by the decreased expression of PRC1 in EndoC-βH1 cells within the network related to apoptosis of islets of Langerhans (A) and the quantity of pancreatic cells (B) through IPA. Cell viability measurement (C) performed at baseline (100%) and 72 h post transfection of siRNA (% compared to baseline). Data at 72 h represent mean values ± SEM of at least five independent experiments. ***p < 0.001.

References

    1. Bonnefond A., Froguel P. Rare and common genetic events in type 2 diabetes: what should biologists know? Cell Metabolism. 2015;21(3):357–368.
    1. Marullo L., El-Sayed Moustafa J.S., Prokopenko I. Insights into the genetic susceptibility to type 2 diabetes from genome-wide association studies of glycaemic traits. Current Diabetes Reports. 2014;14(11):551.
    1. Pasquali L., Gaulton K.J., Rodríguez-Seguí S.A., Mularoni L., Miguel-Escalada I., Akerman I. Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nature Genetics. 2014;46(2):136–143.
    1. Warren H.R., Evangelou E., Cabrera C.P., Gao H., Ren M., Mifsud B. Genome-wide association analysis identifies novel blood pressure loci and offers biological insights into cardiovascular risk. Nature Genetics Advance Online Publication. 2017
    1. Bonnefond A., Lomberk G., Buttar N., Busiah K., Vaillant E., Lobbens S. Disruption of a novel Kruppel-like transcription factor p300-regulated pathway for insulin biosynthesis revealed by studies of the c.-331 INS mutation found in neonatal diabetes mellitus. The Journal of Biological Chemistry. 2011;286(32):28414–28424.
    1. Martino L., Masini M., Bugliani M., Marselli L., Suleiman M., Boggi U. Mast cells infiltrate pancreatic islets in human type 1 diabetes. Diabetologia. 2015;58(11):2554–2562.
    1. Marselli L., Thorne J., Dahiya S., Sgroi D.C., Sharma A., Bonner-Weir S. Gene expression profiles of beta-cell enriched tissue obtained by laser capture microdissection from subjects with type 2 diabetes. PLoS One. 2010;5(7):e11499.
    1. Lukowiak B., Vandewalle B., Riachy R., Kerr-Conte J., Gmyr V., Belaich S. Identification and purification of functional human-cells by a new specific zinc-fluorescent probe. Journal of Histochemistry & Cytochemistry. 2001;49(4):519–527.
    1. Eisenberg E., Levanon E.Y. Human housekeeping genes, revisited. Trends in Genetics: TIG. 2013;29(10):569–574.
    1. Melé M., Ferreira P.G., Reverter F., DeLuca D.S., Monlong J., Sammeth M. The human transcriptome across tissues and individuals. Science. 2015;348(6235):660–665.
    1. Wu C., Orozco C., Boyer J., Leglise M., Goodale J., Batalov S. BioGPS: an extensible and customizable portal for querying and organizing gene annotation resources. Genome Biology. 2009;10(11):R130.
    1. Ravassard P., Hazhouz Y., Pechberty S., Bricout-Neveu E., Armanet M., Czernichow P. A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion. Journal of Clinical Investigation. 2011;121(9):3589–3597.
    1. Bonner C., Kerr-Conte J., Gmyr V., Queniat G., Moerman E., Thévenet J. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nature Medicine. 2015;21(5):512–517.
    1. Kim D., Pertea G., Trapnell C., Pimentel H., Kelley R., Salzberg S.L. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biology. 2013;14(4):R36.
    1. Liao Y., Smyth G.K., Shi W. The subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Research. 2013;41(10):e108.
    1. Love M.I., Huber W., Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology. 2014;15(12):550.
    1. Postic C., Shiota M., Niswender K.D., Jetton T.L., Chen Y., Moates J.M. Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase. The Journal of Biological Chemistry. 1999;274(1):305–315.
    1. Miura A., Yamagata K., Kakei M., Hatakeyama H., Takahashi N., Fukui K. Hepatocyte nuclear factor-4alpha is essential for glucose-stimulated insulin secretion by pancreatic beta-cells. The Journal of Biological Chemistry. 2006;281(8):5246–5257.
    1. Bouzakri K., Ribaux P., Tomas A., Parnaud G., Rickenbach K., Halban P.A. Rab GTPase-activating protein AS160 is a major downstream effector of protein kinase B/Akt signaling in pancreatic-cells. Diabetes. 2008;57(5):1195–1204.
    1. Helman A., Klochendler A., Azazmeh N., Gabai Y., Horwitz E., Anzi S. p16(Ink4a)-induced senescence of pancreatic beta cells enhances insulin secretion. Nature Medicine. 2016;22(4):412–420.
    1. Krautkramer K.A., Linnemann A.K., Fontaine D.A., Whillock A.L., Harris T.W., Schleis G.J. Tcf19 is a novel islet factor necessary for proliferation and survival in the INS-1-cell line. AJP: Endocrinology and Metabolism. 2013;305(5):E600–E610.
    1. Mitchell R.K., Hu M., Chabosseau P.L., Cane M.C., Meur G., Bellomo E.A. Molecular genetic regulation of Slc30a8/ZnT8 reveals a positive association with glucose tolerance. Molecular Endocrinology. 2016;30(1):77–91.
    1. Vierra N.C., Dadi P.K., Jeong I., Dickerson M., Powell D.R., Jacobson D.A. The type-2 diabetes-associated K+ channel TALK-1 modulates beta-cell electrical excitability, 2nd-phase insulin secretion, and glucose homeostasis. Diabetes. 2015
    1. Rutter G.A., Chimienti F. SLC30A8 mutations in type 2 diabetes. Diabetologia. 2015;58(1):31–36.
    1. Sladek R., Rocheleau G., Rung J., Dina C., Shen L., Serre D. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature. 2007;445(7130):881–885.
    1. Kim I., Kang E.S., Yim Y.S., Ko S.J., Jeong S.-H., Rim J.H. A low-risk ZnT-8 allele (W325) for post-transplantation diabetes mellitus is protective against cyclosporin A-induced impairment of insulin secretion. The Pharmacogenomics Journal. 2011;11(3):191–198.
    1. Nicolson T.J., Bellomo E.A., Wijesekara N., Loder M.K., Baldwin J.M., Gyulkhandanyan A.V. Insulin storage and glucose homeostasis in mice null for the granule zinc transporter ZnT8 and studies of the type 2 diabetes-associated variants. Diabetes. 2009;58(9):2070–2083.
    1. Flannick J., Thorleifsson G., Beer N.L., Jacobs S.B.R., Grarup N., Burtt N.P. Loss-of-function mutations in SLC30A8 protect against type 2 diabetes. Nature Genetics. 2014;46(4):357–363.
    1. Sano H., Kane S., Sano E., Mîinea C.P., Asara J.M., Lane W.S. Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. The Journal of Biological Chemistry. 2003;278(17):14599–14602.
    1. Moltke I., Grarup N., Jørgensen M.E., Bjerregaard P., Treebak J.T., Fumagalli M. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature. 2014;512(7513):190–193.
    1. Pal A., Potjer T.P., Thomsen S.K., Ng H.J., Barrett A., Scharfmann R. Loss-of-function mutations in the cell-cycle control gene CDKN2A impact on glucose homeostasis in humans. Diabetes. 2016;65(2):527–533.
    1. Thomsen S.K., Ceroni A., van de Bunt M., Burrows C., Barrett A., Scharfmann R. Systematic functional characterization of candidate causal genes for type 2 diabetes risk variants. Diabetes. 2016;65(12):3805–3811.
    1. Chen J., Rajasekaran M., Xia H., Zhang X., Kong S.N., Sekar K. The microtubule-associated protein PRC1 promotes early recurrence of hepatocellular carcinoma in association with the Wnt/β-catenin signalling pathway. Gut. 2016;65(9):1522–1534.
    1. Zhao Z., Wen W., Michailidou K., Bolla M.K., Wang Q., Zhang B. Association of genetic susceptibility variants for type 2 diabetes with breast cancer risk in women of European ancestry. Cancer Causes & Control. 2016:1–15.
    1. Hagiwara H., Iyo M., Hashimoto K. Neonatal disruption of serine racemase causes schizophrenia-like behavioral abnormalities in adulthood: clinical rescue by d-serine. PLoS One. 2013;8(4):e62438.
    1. Cobb J., Eckhart A., Perichon R., Wulff J., Mitchell M., Adam K.-P. A novel test for IGT utilizing metabolite markers of glucose tolerance. Journal of Diabetes Science and Technology. 2015;9(1):69–76.
    1. Bentley-Lewis R., Huynh J., Xiong G., Lee H., Wenger J., Clish C. Metabolomic profiling in the prediction of gestational diabetes mellitus. Diabetologia. 2015;58(6):1329–1332.
    1. Walter P., Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011;334(6059):1081–1086.

Source: PubMed

3
Sottoscrivi