Mitochondrial glucose metabolism is essential for stimulated insulin release from pancreatic β-cells. Whether mitofusin gene expression, and hence, mitochondrial network integrity, is important for glucose or incretin signaling has not previously been explored. Here, we generated mice with β-cell–selective, adult-restricted deletion knock-out (dKO) of the mitofusin genes Mfn1 and Mfn2Mfn1/2 dKO). βMfn1/2-dKO mice displayed elevated fed and fasted glycemia and a more than fivefold decrease in plasma insulin. Mitochondrial length, glucose-induced polarization, ATP synthesis, and cytosolic and mitochondrial Ca2+ increases were all reduced in dKO islets. In contrast, oral glucose tolerance was more modestly affected in βMfn1/2-dKO mice, and glucagon-like peptide 1 or glucose-dependent insulinotropic peptide receptor agonists largely corrected defective glucose-stimulated insulin secretion through enhanced EPAC-dependent signaling. Correspondingly, cAMP increases in the cytosol, as measured with an Epac-camps–based sensor, were exaggerated in dKO mice. Mitochondrial fusion and fission cycles are thus essential in the β-cell to maintain normal glucose, but not incretin, sensing. These findings broaden our understanding of the roles of mitofusins in β-cells, the potential contributions of altered mitochondrial dynamics to diabetes development, and the impact of incretins on this process.

Mitochondria are often referred to as the powerhouses or “chief executive organelles” of the cell, using fuels to provide most of the energy required to sustain normal function (1). Mitochondrial oxidative metabolism plays a pivotal role in the response of pancreatic β-cells to stimulation by glucose and other nutrients (2). Thus, as blood glucose increases, enhanced glycolytic flux and oxidative metabolism lead to an increase in ATP synthesis, initiating a cascade of events that involve the closure of KATP channels (3), plasma membrane depolarization, and the influx of Ca2+ via voltage-dependent Ca2+ channels (VDCC). The latter, along with other less well-defined “amplifying” signals (4), drive the biphasic release of insulin (2). Gut-derived incretin hormones, including glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) (5), further potentiate secretion by binding to class-B G protein-coupled receptors (GPCRs) to generate cAMP and other intracellular signals (5).

Under normal physiological conditions, mitochondria undergo fusion and fission cycles that are essential for quality control and adaptation to energetic demands (6). Thus, highly interconnected mitochondrial networks allow communication and interchange of contents between mitochondrial compartments as well as with other organelles such as the endoplasmic reticulum (ER) (7). These networks exist interchangeably with more fragmented structures, displaying more “classical” mitochondrial morphology (8). Mitochondrial fission is also necessary for “quality control” and the elimination of damaged mitochondria by mitophagy (9).

While the mitofusins MFN1 and MFN2, homologs of the Drosophila melanogaster fuzzy onions (fzo) and mitofusin (dmfn) gene products (10), are guanosine-5'-triphosphatases that mediate fusion of the outer mitochondrial membrane, optic atrophy protein 1 (OPA1) controls that of the inner mitochondrial membrane. Dynamin-related protein 1 (DRP1) is responsible for mitochondrial fission (11). Other regulators include mitochondrial fission 1 protein (FIS1), mitochondrial fission factor (MFF), and MiD49/51 (12).

Earlier studies (1318) have shown that perturbations in mitochondrial structure in β-cells have marked effects on glucose-stimulated insulin secretion (GSIS). Surprisingly, whether the canonical and evolutionarily conserved machinery involved in mitochondrial fusion (i.e., the mitofusins), control mitochondrial structure in β-cells has not been explored yet. Furthermore, none of the earlier studies have investigated the actions of mitochondrial structure destruction in adult mice. Finally, whether and to what extent they impact secretion stimulated by other agents, including incretins, is less clear. This question is important given that changes in mitochondrial oxidative metabolism (19) and structure contribute to type 2 diabetes (T2D).

Here, we first explored the potential contribution of mitofusins to the effects of diabetic conditions. We next determined whether deletion of Mfn1 and Mfn2 in β-cells in adult mice may impact insulin secretion. Lastly, we aimed to determine whether incretins may rescue or bypass any observed perturbations. We show that mitofusin ablation exerts profound effects on insulin release, glucose homeostasis, and Ca2+ dynamics. Remarkably, the deficiencies in insulin secretion are largely corrected by incretin hormones. This suggests a possible approach to ameliorating the consequences of mitochondrial fragmentation with these agonists in some forms of diabetes.

Study Approval

C57BL/6J mice were housed in individually ventilated cages in a pathogen-free facility at 22°C with a 10–14-h light-dark cycle and were fed ad libitum with a standard mouse chow diet (Research Diets, New Brunswick, NJ). All in vivo procedures were approved by the U.K. Home Office, according to the Animals (Scientific Procedures) Act 1986 with local ethical committee (Hammersmith Hospital Campus, London, U.K.) approval under personal project license number PA03F7F07 to I.L.

Generation of β-Cell Selective Mfn1/Mfn2 Knockout, Clec16a Null, and Pdx1CreER Mice

C57BL/6J male mice bearing Mfn1 (Mfn1tm2Dcc; JAX stock no. 026401) and Mfn2 (B6.129(Cg)-Mfn2tm3Dcc/J; JAX stock no. 026525; The Jackson Laboratory, Bar Harbor, ME) alleles (20) with loxP sites flanking exons 4 and 6 were purchased from The Jackson Laboratory and crossed to C57BL/6J transgenic animals carrying an inducible Cre recombinase under Pdx1 promoter control (Pdx1-CreERT2) (21). Mice bearing floxed Mfn alleles but lacking Cre recombinase were used as littermate controls in this study. Mice were genotyped following protocols described by The Jackson Laboratory for each of these strains (see Supplementary Table 1). Recombination was achieved by daily tamoxifen (10 mg/mouse diluted in corn oil; Sigma-Aldrich, Dorset, U.K.) i.p. injections for 5 days at 7–8 weeks of age in both control and β-cell selective Mfn1/Mfn2 deletion knockout (dKO) (βMfn1/2 dKO) groups.

Animals with floxed Clec16a alleles were bred to mice carrying the Pdx1-Cre transgene (Clec16aΔpanc), as previously described (22). Pdx1-Cre alone mice were used as littermate controls. Pdx1CreER mice were generated as previously described (21).

RNA Extraction and Quantitative RT-PCR

For measurements of mRNA levels, pancreatic islets from control and βMfn1/2-dKO mice were isolated by collagenase digestion (23). Total RNA from 50 to 100 islets was extracted and underwent quantitative (q)RT-PCR, as previously described (24) (see Supplementary Table 2 for primer details).

Tissue DNA Extraction and Measurement of mtDNA Copy Number

Total islet DNA was isolated using Puregene Cell and Tissue Kit (Qiagen, Manchester, U.K.) and was amplified (100 ng) using NADH dehydrogenase I primers (25), also known as complex I (mt9/mt11) for mtDNA and Ndufv1 for nuclear DNA.

SDS-PAGE and Western Blotting

Islets were collected and lysed (20 μg), as previously described (24). The antibodies used are summarized in Supplementary Table 3.

Intraperitoneal or Oral Gavage of Glucose, Followed by Insulin, Proinsulin, or Ketone Levels Measurement and Insulin Tolerance Test In Vivo

Intraperitonal glucose tolerance tests (IPGTTs), intraperitoneal insulin tolerance tests (IPIITTs), oral glucose tolerance tests (OGTTs), and plasma insulin measurements were performed as previously described (24). Plasma proinsulin levels were measured in fasted (16 h) animals using a rat/mouse proinsulin ELISA kit (Mercodia). Plasma β-ketones were measured from fed or fasted (16 h) mice using an Areo 2K device (GlucoMen, Berkshire, U.K.).

In Vitro Insulin Secretion

Islets were isolated from mice and incubated for 1 h in Krebs-Ringer bicarbonate buffer containing 3 mmol/L glucose, as previously described (24).

Single-Cell Fluorescence Imaging

Dissociated islets were incubated with 100 nmol/L MitoTracker Green (Thermo Fisher Scientific) in Krebs-Ringer bicarbonate buffer containing 11 mmol/L glucose for 30 min. MitoTracker Green was then washed with Krebs buffer with 11 mmol/L glucose before fluorescence imaging. Experiments with tetramethylrhodamine ethyl ester (TMRE) were performed as previously described (24). Clusters of dissociated islets were transduced for 48 h with an adenovirus encoding the low-Ca2+-affinity sensor D4 addressed to the ER, Ad-RIP-D4ER (multiplicity of infection: 100), as previously described (26). Bleaching was corrected as previously described (27). Clusters of dissociated islets were transduced for 24 h with an adenovirus encoding Epac1-camps, as previously described (28).

Mitochondrial Shape Analysis

For each stack, one image at the top, middle, and bottom of the islet was analyzed. After background subtraction, the following parameters were measured for each cell: number of particles, perimeter, circularity, elongation (1/circularity), density, and surface area of each particle (29).

Whole-Islet Fluorescence Imaging

Cytosolic and mitochondrial Ca2+ imaging as well as ATP-to-ADP changes in whole islets were performed as previously described (24).

TIRF Fluorescence Imaging

Experiments using the membrane-located zinc sensor ZIMIR (50 μmol/L) (30) or the fluorescent genetically encoded and vesicle-located green marker neuropeptide Y (NPY)-Venus were performed as previously described (31).

Pancreas Immunohistochemistry

Isolated pancreata were fixed and imaged as previously described (24). The antibodies used are summarized in Supplementary Table 3. For examination of apoptosis, TUNEL assay was performed using a DeadEnd Fluorometric TUNEL system kit and DNase I treatment (Promega, Madison, WI), according to the manufacturer's instructions.

Metabolomics/Lipidomics

Metabolites were quantified using targeted ultrahigh-performance liquid chromatography coupled with triple quadrupole mass spectrometry, as described earlier (32). Lipidomic sample preparation followed the Folch procedure with minor adjustments. Significance was tested by the Student two-tailed t test using GraphPad Prism 8 software.

Measurement of Oxygen Consumption Rate

The oxygen consumption rate (OCR) was measured with XF96 assays (Seahorse Bioscience, Agilent, Santa Clara, CA) using mouse islets (∼10 per well), as previously described (33). Parameters were analyzed as previously described (34).

Electron Microscopy

For conventional electron microscopy, islets were fixed and imaged as previously described (35).

Connectivity Analysis

Pearson (r)-Based Connectivity and Correlation Analyses

Correlation analyses in an imaged islet were performed as previously described (36).

RNA Sequencing Data Analysis

Processing and differential expression analysis of RNA sequencing data from islets isolated from mice fed a high-fat high-sugar diet (HFHS; D12331, Research Diets) or regular chow (RC) diet (C57Bl/6J, DBA/2J, BALB/cJ, A/J, AKR/J, 129S2/SvPas) was performed as previously described (37) using the Limma package in R. P values were adjusted for multiple comparisons using the Benjamini-Hochberg procedure (38).

Statistics

Data are expressed as mean ± SD, unless otherwise stated. Significance was tested by the Student two-tailed t test and Mann-Whitney correction or two-way ANOVA with the Šidák multiple comparison test for comparison of more than two groups, using GraphPad Prism 9 software (GraphPad Software, San Diego, CA). P <  0.05 was considered significant. Experiments were not randomized or blinded.

Data and Resource Availability

The data sets generated and/or analyzed during the current study are available from the corresponding author upon reasonable request. No applicable resources were generated or analyzed during the current study.

Changes in Mfn1 and Mfn2 Expression in Mouse Strains Maintained on RC or HFHS Diet

To determine whether the expression of Mfn1 or Mfn2 might be affected under conditions of hyperglycemia mimicking T2D in humans, we interrogated data from a previous report (37) in which RNA sequencing was performed on six mouse strains. BALB/cJ mice showed “antiparallel” changes in Mfn1 and Mfn2 expression in response to maintenance on an HFHS diet for 10 days, and similar changes were obtained in DBA/2J mice at 30 and 90 days (Supplementary Fig. 1A and B).

Generation of a Conditional βMfn1/2-dKO Mouse Line

Efficient deletion of Mfn1 and Mfn2 in the β-cell was achieved in adult mice using the Pdx1-CreERT2 transgene and tamoxifen injection at 7–8 weeks. Possession of this transgene, which does not contain the human growth hormone (hGH) cDNA (21), alone had no effect on glycemic phenotype or cellular composition of pancreatic islets (Supplementary Fig. 2AC). Deletion of mitofusin genes was confirmed by qRT-PCR (Fig. 1A) and Western (immuno-) blotting (Fig. 1B) analysis, ∼7 weeks posttamoxifen injection. Relative to β-actin, expression of the Mfn1 and Mfn2 transcripts in isolated islets from dKO mice decreased by ∼ 83 and 86% accordingly versus control islets (Fig. 1A), consistent with selective deletion in the β-cell compartment (39). No differences were detected in the expression of other mitochondrial fission and fusion mediator genes such as Opa1, Drp1, and Fis1 in islets (Fig. 1A) or in Mfn1 and Mfn2 in other relevant tissues (Supplementary Fig. 3A). dKO mice were significantly lighter than control animals after 20–21 weeks (Supplementary Fig. 3B).

Figure 1

Generation of a conditional βMfn1/2-dKO mouse line, which displays a highly impaired glucose tolerance in vivo. A: qRT-PCR quantification of Mfn1, Mfn2, Drp1, Opa1, and Fis1 expression in control and dKO islets relative to β-actin (n = 3–5 mice per genotype in two independent experiments). B: Western blot analysis demonstrating efficient MFN1 (84 kDa) and MFN2 (86 kDa) deletion relative to GAPDH (36 kDa) in isolated islets (n = 3–4 mice per genotype in three independent experiments). C: Glucose tolerance was measured in dKO mice and littermate controls by IPGTT (1 g/kg body wt). D: Corresponding area under the curve (AUC; AU, arbitrary units) from (C) (n = 8 mice per genotype, in two independent experiments). Glucose tolerance measured by IPGTT (using 3 g/kg body wt) (E), and the corresponding AUC (F) were assessed in βMfn1/2-dKO and control mice (n = 8 mice per genotype in two independent experiments). Plasma insulin levels during IPGTT in dKO and control mice (n = 11–12 mice per genotype in three independent experiments) (G), and the corresponding AUC (H). I: Glucose tolerance after oral gavage (3 g/kg body wt) was measured in eight animals per genotype in two independent experiments. Glucose baseline values between control and dKO mice were significantly different (*P < 0.05). Increases in glucose from baseline in control animals were ****P < 0.0001, ***P < 0.001, and **P < 0.01 and were ****P < 0.0001 in dKO animals from 15 to 60 min accordingly. J: The corresponding AUC. Plasma insulin levels during OGTT in dKO and control mice (n = 8 mice per genotype in two independent experiments) (K) and the corresponding AUC (L). All experiments were performed in 14-week-old male mice. Data are presented as mean ± SD in A and as mean ± SEM in BL. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001, as indicated, or control vs. dKO mice at the time points as indicated in K, analyzed by unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Figure 1

Generation of a conditional βMfn1/2-dKO mouse line, which displays a highly impaired glucose tolerance in vivo. A: qRT-PCR quantification of Mfn1, Mfn2, Drp1, Opa1, and Fis1 expression in control and dKO islets relative to β-actin (n = 3–5 mice per genotype in two independent experiments). B: Western blot analysis demonstrating efficient MFN1 (84 kDa) and MFN2 (86 kDa) deletion relative to GAPDH (36 kDa) in isolated islets (n = 3–4 mice per genotype in three independent experiments). C: Glucose tolerance was measured in dKO mice and littermate controls by IPGTT (1 g/kg body wt). D: Corresponding area under the curve (AUC; AU, arbitrary units) from (C) (n = 8 mice per genotype, in two independent experiments). Glucose tolerance measured by IPGTT (using 3 g/kg body wt) (E), and the corresponding AUC (F) were assessed in βMfn1/2-dKO and control mice (n = 8 mice per genotype in two independent experiments). Plasma insulin levels during IPGTT in dKO and control mice (n = 11–12 mice per genotype in three independent experiments) (G), and the corresponding AUC (H). I: Glucose tolerance after oral gavage (3 g/kg body wt) was measured in eight animals per genotype in two independent experiments. Glucose baseline values between control and dKO mice were significantly different (*P < 0.05). Increases in glucose from baseline in control animals were ****P < 0.0001, ***P < 0.001, and **P < 0.01 and were ****P < 0.0001 in dKO animals from 15 to 60 min accordingly. J: The corresponding AUC. Plasma insulin levels during OGTT in dKO and control mice (n = 8 mice per genotype in two independent experiments) (K) and the corresponding AUC (L). All experiments were performed in 14-week-old male mice. Data are presented as mean ± SD in A and as mean ± SEM in BL. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001, as indicated, or control vs. dKO mice at the time points as indicated in K, analyzed by unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Close modal

βMfn1/2-dKO Mice Are Glucose Intolerant With Impaired GSIS In Vivo

Glucose tolerance was impaired in dKO mice compared with control littermates at 14 weeks (Fig. 1C and D), and this difference was further exaggerated at 20 weeks (Supplementary Fig. 3 and Fig. 3C). At 14 weeks, βMfn1/2-dKO mice (with a 27 mmol/L glycemia at 15 min) (Fig. 1E and F) showed a dramatically lower insulin excursion upon glucose challenge versus control animals (Fig 1G and H). Following an oral gavage, glucose tolerance was more modestly affected in dKO mice (Fig. 1I and J), while plasma insulin levels in these animals (with a glycemia of 27 mmol/L at 15 min) were indistinguishable from control animals (0 vs. 15 min in dKO) (Fig. 1K and L). Insulin tolerance was unaltered in βMfn1/2-dKO versus control mice (Supplementary Fig. 3D), while proinsulin conversion was impaired (Supplementary Fig. 3E and F). dKO mice displayed significantly elevated plasma glucose (Supplementary Fig. 3G) under both fed and fasted conditions, and β-ketones (ketone bodies) were also elevated in fasted versus control animals (Supplementary Fig. 3H), whereas plasma insulin levels were lower (Supplementary Fig. 3I). Apparent insulin secretion was also impaired after i.p. injection, with a lower glucose in 14- and 20-week-old dKO versus control mice (Supplementary Fig. 4AD). In contrast, plasma insulin levels were not statistically different between control and dKO animals following an OGTT at either age (Supplementary Fig. 4EH), although a trend toward lower insulin excursion was evident in dKO mice.

Deletion of Mfn1/2 Alters Mitochondrial Morphology in β-Cells

While the mitochondrial network was highly fragmented in dKO cells (Fig. 2A and inset), the number of mitochondria per cell or density was not altered (Fig. 2B). Mitochondrial elongation, perimeter, and surface area were also significantly decreased in βMfn1/2-dKO cells, while circularity was increased (Fig. 2B). Transmission electron microscopy confirmed these changes (Fig. 2C). Cristae structure and organization were also altered in βMfn1/2-dKO cells, with a single crista often running the length of a mitochondrial section. Finally, dKO islets displayed an ∼5% reduction in mtDNA (Fig. 2D).

Figure 2

Mitochondrial ultrastructure is altered following Mfn1/2 deletion. A: Confocal images of the mitochondrial network of dissociated β-cells stained with MitoTracker Green; scale bar: 5 μm. Lower right panels: magnification of selected areas. B: Mitochondrial morphology analysis on deconvolved confocal images of dissociated β-cells. A macro was developed to quantify the number of mitochondria per cell and measure the elongation, perimeter, circularity (0: elongated; 1: circular mitochondria), density, and surface area of the organelles in control and dKO animals (n = 40–54 cells; n = 3 mice per genotype). C: Electron micrographs of mitochondria indicated with black arrows in islets isolated from control and dKO mice; scale bars: 1 μm. Right panel: magnification of selected areas showing the cristae structure (black arrow heads); scale bar: 0.5 μm. Schematic representation of enlarged mitochondria. D: The relative mitochondrial DNA copy number was measured by determining the ratio of the mtDNA-encoded gene mt-Nd1 to the nuclear gene Ndufv1 (n = 3 mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SEM in AC and as mean ± SD in D. *P < 0.05, ***P < 0.001, ****P < 0.0001 as indicated, analyzed by unpaired two-tailed Student t test and Mann-Whitney correction.

Figure 2

Mitochondrial ultrastructure is altered following Mfn1/2 deletion. A: Confocal images of the mitochondrial network of dissociated β-cells stained with MitoTracker Green; scale bar: 5 μm. Lower right panels: magnification of selected areas. B: Mitochondrial morphology analysis on deconvolved confocal images of dissociated β-cells. A macro was developed to quantify the number of mitochondria per cell and measure the elongation, perimeter, circularity (0: elongated; 1: circular mitochondria), density, and surface area of the organelles in control and dKO animals (n = 40–54 cells; n = 3 mice per genotype). C: Electron micrographs of mitochondria indicated with black arrows in islets isolated from control and dKO mice; scale bars: 1 μm. Right panel: magnification of selected areas showing the cristae structure (black arrow heads); scale bar: 0.5 μm. Schematic representation of enlarged mitochondria. D: The relative mitochondrial DNA copy number was measured by determining the ratio of the mtDNA-encoded gene mt-Nd1 to the nuclear gene Ndufv1 (n = 3 mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SEM in AC and as mean ± SD in D. *P < 0.05, ***P < 0.001, ****P < 0.0001 as indicated, analyzed by unpaired two-tailed Student t test and Mann-Whitney correction.

Close modal

Mitofusin Deletion Leads to Modest Changes in β-Cell Mass

Pancreatic β-cell mass decreased by 33%, whereas α-cell mass was not affected in dKO mice (Fig. 3A–C). The β-cell–to–α-cell ratio was decreased by 53% (Fig. 3D), in line with an increase in TUNEL-positive β-cells in dKO versus control animals (Fig. 3E and F).

Figure 3

Absence of Mfn1/2 in β-cells leads to decreased β-cell mass and increased β-cell apoptosis. A: Representative pancreatic sections immunostained with glucagon (red) and insulin (green); scale bars: 50 μm. The β-cell and α-cell surface (B) measured within the whole pancreatic area in control and dKO mice were determined (C), as well as the β-cell–to–α-cell ratio in D (n = 79–86 islets, four mice per genotype; experiment performed in triplicate). E: Representative confocal images of islets with TUNEL-positive (green) apoptotic β-cells (region of interest) and insulin (red). Magnification of selected area displaying each fluorescent channel; scale bar: 5 μm. DNase I-treated sections were used as a positive control in the TUNEL assay. Scale bars: 20 μm. F: Quantification of the percentage of islets containing TUNEL-positive cells (n = 114–133 islets, four mice per genotype; experiment performed in triplicate). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, assessed by unpaired two-tailed Student t test and Mann-Whitney correction.

Figure 3

Absence of Mfn1/2 in β-cells leads to decreased β-cell mass and increased β-cell apoptosis. A: Representative pancreatic sections immunostained with glucagon (red) and insulin (green); scale bars: 50 μm. The β-cell and α-cell surface (B) measured within the whole pancreatic area in control and dKO mice were determined (C), as well as the β-cell–to–α-cell ratio in D (n = 79–86 islets, four mice per genotype; experiment performed in triplicate). E: Representative confocal images of islets with TUNEL-positive (green) apoptotic β-cells (region of interest) and insulin (red). Magnification of selected area displaying each fluorescent channel; scale bar: 5 μm. DNase I-treated sections were used as a positive control in the TUNEL assay. Scale bars: 20 μm. F: Quantification of the percentage of islets containing TUNEL-positive cells (n = 114–133 islets, four mice per genotype; experiment performed in triplicate). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, assessed by unpaired two-tailed Student t test and Mann-Whitney correction.

Close modal

Mitochondrial Fragmentation, β-Cell Mass Deterioration, and Hyperglycemia Emerge in dKO Mice 2 Weeks After Tamoxifen Administration

We next sought to exclude the possibility that mitochondrial fragmentation may simply be the consequence of the observed hyperglycemia. Two distinct groups of organelles (both elongated and circular) were apparent in βMfn1/2-dKO cells (Supplementary Fig. 5A and B) 2 weeks after tamoxifen treatment. Neither fed nor fasted glycemia or plasma insulin levels following glucose challenge were different between groups (Supplementary Fig. 5CE). A trend toward lower β-cell mass and mtDNA was detected in dKO animals (Supplementary Fig. 5F–I).

β-Cell Identity Is Modestly Altered in βMfn1/2-dKO Islets

While Ins2, Ucn3, and Glut2 (Slc2a2) were significantly downregulated, Trpm5 was upregulated in dKO islets (Supplementary Fig. 6). No changes in α- or β-cell disallowed genes (40) were detected. In contrast, genes involved in mitochondrial function, such as Smdt1 and Vdac3, were upregulated in dKO β-cells (Supplementary Fig. 6). Lastly, genes involved in ER stress and mito/autophagy were also affected, with Chop (Ddit3) and p62 being upregulated and Lc3 and Cathepsin L downregulated.

Mitofusins Are Essential to Maintain Normal Glucose-Stimulated Ca2+ Dynamics, Mitochondrial Membrane Potential, and ATP Levels

Increased cytosolic Ca2+ is a key trigger of insulin exocytosis in response to high glucose (2). dKO mouse islets exhibited a significantly smaller glucose-induced [Ca2+]cyt rise versus control islets (Fig. 4A–C). When the KATP channel opener diazoxide and a depolarizing K+ concentration were then deployed together to bypass the regulation of these channels by glucose, cytosolic Ca2+ increases were not significantly impaired in dKO compared with control animals (Fig. 4B and C). A substantial reduction in mitochondrial free Ca2+ concentration ([Ca2+]mito) in response to 17 mmol/L glucose (24) was also observed in dKO islets (Fig. 4D–F). Of note, subsequent hyperpolarization of the plasma membrane with diazoxide caused the expected lowering of mitochondrial [Ca2+]mito in control islets, reflecting the decrease in [Ca2+]cyt (Fig. 4E and F), but was almost without effect on dKO islets.

Figure 4

Mfn1/2 deletion from pancreatic β-cells impairs cytosolic and mitochondrial Ca2+ uptake and changes mitochondrial potential and ATP synthesis in vitro. A: Each snapshot of isolated control (i–iv) and dKO-derived (v–viii) islets was taken during the time points indicated by the respective arrows in B. Scale bar: 50 μm. See also Supplementary Video 1. B: [Ca2+]cyt traces in response to 3 mmol/L glucose (3G), 17 mmol/L glucose (17G), with or without 100 μmol/L diazoxide (diaz), or 20 mmol/L KCl with diaz were assessed following Cal-520 uptake in whole islets. Traces represent mean normalized fluorescence intensity over time (F/Fmin). C: The corresponding area under the curve (AUC) is also presented (AU, arbitrary units) (n = 17–26 islets, four mice per genotype); 17G AUC was measured between 245 s and 1,045 s, 17G+diaz AUC was measured between 1,200 s and 1,320 s, and KCl+diaz AUC was measured between 1,424 s and 1,500 s. For each genotype, different baselines (ctrl diaz/KCl: 0.95, dKO diaz/KCl: 0.8) were taken into consideration to measure AUCs. D: Each snapshot of isolated control (i–iv) and dKO-derived (v–viii) islets was taken during the time points indicated by the respective arrows in E. Scale bar: 50 μm. See also Supplementary Video 2. E: [Ca2+]mito changes in response to 17G, with or without diaz, and 20 mmol/L KCl were assessed in islets following R-GECO infection. Traces represent mean normalized fluorescence intensity over time (F/Fmin), where Fmin is the mean fluorescence recorded during imaging under 3G. F: The corresponding AUC is also shown (n = 20–23 islets, three mice per genotype); 17G AUC was measured between 270 s and 1,100 s, 17G+diaz AUC was measured between 1,101 s and 1,365 s, and KCl AUC was measured between 1,366 s and 1,500 s. G: Dissociated β-cells were loaded with TMRE to measure changes in Δψm and perifused with 3G, 17G, or FCCP, as indicated. Traces represent normalized fluorescence intensity over time (F/Fmin). H: AUC was measured between 700 s and 730 s (under 17G exposure) from the data shown in G (n = 146–254 cells, three to six mice per genotype). I: Changes in the cytoplasmic ATP-to-ADP ratio in response to 17G was examined in whole islets using the ATP sensor Perceval. J: AUC values corresponding to I were measured between 418 s and 1,400 s (under 17G exposure) (data points from n = 22–23 islets, three to six mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, assessed by unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Figure 4

Mfn1/2 deletion from pancreatic β-cells impairs cytosolic and mitochondrial Ca2+ uptake and changes mitochondrial potential and ATP synthesis in vitro. A: Each snapshot of isolated control (i–iv) and dKO-derived (v–viii) islets was taken during the time points indicated by the respective arrows in B. Scale bar: 50 μm. See also Supplementary Video 1. B: [Ca2+]cyt traces in response to 3 mmol/L glucose (3G), 17 mmol/L glucose (17G), with or without 100 μmol/L diazoxide (diaz), or 20 mmol/L KCl with diaz were assessed following Cal-520 uptake in whole islets. Traces represent mean normalized fluorescence intensity over time (F/Fmin). C: The corresponding area under the curve (AUC) is also presented (AU, arbitrary units) (n = 17–26 islets, four mice per genotype); 17G AUC was measured between 245 s and 1,045 s, 17G+diaz AUC was measured between 1,200 s and 1,320 s, and KCl+diaz AUC was measured between 1,424 s and 1,500 s. For each genotype, different baselines (ctrl diaz/KCl: 0.95, dKO diaz/KCl: 0.8) were taken into consideration to measure AUCs. D: Each snapshot of isolated control (i–iv) and dKO-derived (v–viii) islets was taken during the time points indicated by the respective arrows in E. Scale bar: 50 μm. See also Supplementary Video 2. E: [Ca2+]mito changes in response to 17G, with or without diaz, and 20 mmol/L KCl were assessed in islets following R-GECO infection. Traces represent mean normalized fluorescence intensity over time (F/Fmin), where Fmin is the mean fluorescence recorded during imaging under 3G. F: The corresponding AUC is also shown (n = 20–23 islets, three mice per genotype); 17G AUC was measured between 270 s and 1,100 s, 17G+diaz AUC was measured between 1,101 s and 1,365 s, and KCl AUC was measured between 1,366 s and 1,500 s. G: Dissociated β-cells were loaded with TMRE to measure changes in Δψm and perifused with 3G, 17G, or FCCP, as indicated. Traces represent normalized fluorescence intensity over time (F/Fmin). H: AUC was measured between 700 s and 730 s (under 17G exposure) from the data shown in G (n = 146–254 cells, three to six mice per genotype). I: Changes in the cytoplasmic ATP-to-ADP ratio in response to 17G was examined in whole islets using the ATP sensor Perceval. J: AUC values corresponding to I were measured between 418 s and 1,400 s (under 17G exposure) (data points from n = 22–23 islets, three to six mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, assessed by unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Close modal

Glucose-induced increases in Δψm were also sharply reduced in dKO versus control mouse islets (Fig. 4G and H). Addition of 2-[2-[4-(trifluoromethoxy)phenyl]hydrazinylidene]-propanedinitrile (FCCP) resulted in a similar collapse in apparent Δψm in islets from both genotypes (Fig. 4G). Cytosolic Ca2+ oscillations and synchronous Δψm depolarization were also largely abolished in response to glucose in dKO cells when measured by intravital imaging in vivo (41). Finally, to assess whether deletion of Mfn1 and Mfn2 may impact glucose-induced increases in mitochondrial ATP synthesis, we performed real-time fluorescence imaging using Perceval (Fig. 4I and J). While control islets responded with a time-dependent rise in the ATP-to-ADP ratio in response to a step increase in glucose from 3 mmol/L to 17 mmol/L, βMfn1/2-dKO β-cells failed to mount any response (Fig. 4J).

β-Cell–β-Cell Connectivity Is Impaired by Mfn1/2 Ablation

Intercellular connectivity is required in the islet for a full insulin secretory response to glucose (42). To assess this, individual Ca2+ traces recorded from Cal-520–loaded β-cells in mouse islets (Fig. 4A and B) were subjected to correlation (Pearson r) analysis to map cell-to-cell connectivity (Supplementary Fig. 7A). Following perfusion at 17 mmol/L glucose, βMfn1/2-dKO β-cells tended to display an inferior, although not significantly different, coordinated activity than control cells, as assessed by counting the number of coordinated cell pairs (0.94 vs. 0.90 for control vs. dKO, respectively) (Supplementary Fig. 7C). By contrast, β-cells displayed highly coordinated Ca2+ responses upon addition of 20 mmol/L KCl in dKO islets. Similarly, analysis of correlation strength in the same islets revealed significant differences in response to 17 mmol/L glucose between genotypes. In fact, dKO islets had weaker mean β-cell–to–β-cell coordinated activity (0.88 vs. 0.77 for control vs. dKO, respectively; P < 0.05) (Supplementary Fig. 7B and D), indicating that mitofusins affect the strength of connection rather than the number of coordinated β-cell pairs. A tendency toward lower expression of the gap junction gene Cx36/Gjd2 was observed in dKO islets (Supplementary Fig. 7E). β-Cell “hub” and “leader” distributions (43) were also impaired in the dKO group (data not shown; see [41]).

Unaltered ER Ca2+ Mobilization but Decreased Mitochondrial VO2 and mtDNA Depletion in βMfn1/2-dKO Islets

No differences in cytosolic Ca2+ responses between genotypes were observed after agonism at the Gq-coupled metabotropic acetylcholine (Ach) receptor (44,45) (Fig. 5A–C). In contrast, measurements of VO2 revealed that basal, proton leak, and maximal respiratory capacities were significantly impaired in dKO islets (Fig. 5D and E).

Figure 5

VO2 and mtDNA are deleteriously affected when Mfn1/2 are abolished in β-cells, while [Ca2+]ER mobilization remains unchanged. A: Each snapshot of isolated control (i–v) and dKO-derived (vi–x) islets was taken during the time points indicated by the respective arrows in B. Scale bar: 50 μm. See also Supplementary Video 3. B: Changes in [Ca2+]ER were measured in whole islets incubated with Cal-520 and perifused with 17 mmol/L glucose (17G), with or without diazoxide (diaz), 17G with 100 μmol/L acetylcholine (Ach) and diaz, or 20 mmol/L KCl with diaz. C: Area under the curve (AUC) values (AU, arbitrary units) corresponding to B were measured: 17G AUC was measured between 260 s and 740 s, 17G+diaz AUC was measured between 846 s and 1,020 s, 17G+diaz+Ach AUC was measured between 1,021 s and 1,300 s, and KCl AUC was measured between 1,301 s and 1,500 s (n = 29–31 islets, three mice per genotype). D: Representative OCR traces of islets (∼10 per well) were acutely exposed to 20 mmol/L glucose (final concentration), 5 µmol/L oligomycin A (Oligo), 1 µmol/L FCCP, and 5 µmol/L rotenone (Rot) with antimycin A (AA) (performed in n = 7 mice, in two independent experiments). E: Mitochondrial metabolic parameters were extracted from the OCR traces shown in D. Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD in AC and as mean ± SEM in D and E. *P < 0.05,**P < 0.01, assessed by the unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Figure 5

VO2 and mtDNA are deleteriously affected when Mfn1/2 are abolished in β-cells, while [Ca2+]ER mobilization remains unchanged. A: Each snapshot of isolated control (i–v) and dKO-derived (vi–x) islets was taken during the time points indicated by the respective arrows in B. Scale bar: 50 μm. See also Supplementary Video 3. B: Changes in [Ca2+]ER were measured in whole islets incubated with Cal-520 and perifused with 17 mmol/L glucose (17G), with or without diazoxide (diaz), 17G with 100 μmol/L acetylcholine (Ach) and diaz, or 20 mmol/L KCl with diaz. C: Area under the curve (AUC) values (AU, arbitrary units) corresponding to B were measured: 17G AUC was measured between 260 s and 740 s, 17G+diaz AUC was measured between 846 s and 1,020 s, 17G+diaz+Ach AUC was measured between 1,021 s and 1,300 s, and KCl AUC was measured between 1,301 s and 1,500 s (n = 29–31 islets, three mice per genotype). D: Representative OCR traces of islets (∼10 per well) were acutely exposed to 20 mmol/L glucose (final concentration), 5 µmol/L oligomycin A (Oligo), 1 µmol/L FCCP, and 5 µmol/L rotenone (Rot) with antimycin A (AA) (performed in n = 7 mice, in two independent experiments). E: Mitochondrial metabolic parameters were extracted from the OCR traces shown in D. Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD in AC and as mean ± SEM in D and E. *P < 0.05,**P < 0.01, assessed by the unpaired two-tailed Student t test and Mann-Whitney correction or two-way ANOVA test and the Šidák multiple comparisons test.

Close modal

Impaired GSIS In Vitro and β-Cell Connectivity Can Be Rescued by Incretins in βMfn1/2-dKO Mouse Islets

While GSIS was markedly impaired in dKO islets (Fig. 6A and Supplementary Table 4), incretins (GLP-1 or GIP), or the GLP1R agonist exendin-4, at a submaximal concentration of 10 mmol/L glucose, led to a significant potentiation in GSIS in both groups. Consequently, insulin secretion in response to 10 mmol/L glucose was no longer different between control and βMfn1/2-dKO islets after incretin addition (Fig. 6A and B). Moreover, under these conditions, forced increases in intracellular cAMP imposed by the addition of forskolin (FSK) or 3-isobutyl-1-methylxanthine (IBMX), which activate adenylate cyclase (AC) and inhibit phosphodiesterase, respectively, eliminated differences in GSIS between the genotypes (Fig. 6B). No differences in insulin secretion were observed between control and dKO islets after depolarization with KCl.

Figure 6

Impaired insulin secretion can be rescued by GLP-1R agonists in vitro by increasing cytosolic Ca2+ oscillation frequency. A: Insulin secretion measured during serial incubations in batches in 3 mmol/L glucose (3G), 10 mmol/L glucose (10G), 17 mmol/L glucose (17G), 10G supplemented with 100 nmol/L exendin-4 (ex4), GLP-1, GIP, 10 µmol/L FSK, 100 µmol/L IBMX, or 3G with 20 mmol/L KCl (n = 3–7 mice per genotype in two independent experiments; control: 3G vs. ex4, P < 0.05; and dKO: 3G vs. ex4, P < 0.0001; or 3G vs. GLP-1, P < 0.001; or 3G vs. GIP, P < 0.001). B: Glucose tolerance measured by i.p. coinjection of 1 g/kg glucose and 3 nmol/kg ex4 were assessed in βMfn1/2-dKO and control mice (n = 4–5 mice per genotype, dotted lines). C: [Ca2+]cyt changes in response to 3G, 10G, with or without exendin-4 (ex4), or 20 mmol/L KCl were assessed following Cal-520 uptake in whole islets. Traces represent mean normalized fluorescence intensity over time (F/Fmin). See also Supplementary Video 4. Dashed regions of interest represent fluorescent segments of extended time scales. Both control and dKO traces reveal faster oscillatory frequencies in response to exendin-4. D: The corresponding area under the curve (AUC) is also presented (AU, arbitrary units) (n = 19–20 islets, three mice per genotype). The 10G AUC was measured between 200 s and 660 s, 10G+ex4 AUC was measured between 800 s and 950 s, and KCl AUC was measured between 1,200 s and 1,500 s (AUC 10G: control vs. dKO, P = 0.09; AUC control: 10G vs. ex4, P < 0.05; AUC dKO: 10G vs. ex4, P < 0.001; ex4 vs. KCl, P < 0.05). E: Dissociated β-cells were transfected with D4ER to measure changes in [Ca2+]ER, and perifused with 10G, 10G+ex4, or thapsigargin (10G+thapsi), as indicated. Traces represent corrected ratio values postlinear fitting over time. F: AUC was measured between 350 s and 900 s (under 10G+ex4) and between 900 s and 1,300 s (10G+thapsi) from the data shown in E (n = 44–46 cells, four to five mice per genotype). G: Changes in cytoplasmic ATP-to-ADP ratio in response to 10G or 10G with 100 nmol/L ex4 was examined in whole islets. H: AUC values corresponding to G were measured between 185 s and 720 s (under 10G exposure) or between 721 s and 1,200 s (under 10G with ex4) (data points from n = 3 mice per genotype). I: Average OCR values of islets (∼10 per well) that were exposed to 3G or 10G (final concentration), 10G supplemented with ex4, FSK, oligomycin A (Oligo), FCCP, and rotenone (Rot) with antimycin A (AA) (n = 3 mice per genotype; experiment performed in duplicate). J: Insulin secretion measured during serial incubations in batches in 3G or 17G supplemented with 100 μmol/L diazoxide and 30 mmol/L KCl, (n = 3 mice per genotype in two independent experiments). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ****P < 0.0001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Figure 6

Impaired insulin secretion can be rescued by GLP-1R agonists in vitro by increasing cytosolic Ca2+ oscillation frequency. A: Insulin secretion measured during serial incubations in batches in 3 mmol/L glucose (3G), 10 mmol/L glucose (10G), 17 mmol/L glucose (17G), 10G supplemented with 100 nmol/L exendin-4 (ex4), GLP-1, GIP, 10 µmol/L FSK, 100 µmol/L IBMX, or 3G with 20 mmol/L KCl (n = 3–7 mice per genotype in two independent experiments; control: 3G vs. ex4, P < 0.05; and dKO: 3G vs. ex4, P < 0.0001; or 3G vs. GLP-1, P < 0.001; or 3G vs. GIP, P < 0.001). B: Glucose tolerance measured by i.p. coinjection of 1 g/kg glucose and 3 nmol/kg ex4 were assessed in βMfn1/2-dKO and control mice (n = 4–5 mice per genotype, dotted lines). C: [Ca2+]cyt changes in response to 3G, 10G, with or without exendin-4 (ex4), or 20 mmol/L KCl were assessed following Cal-520 uptake in whole islets. Traces represent mean normalized fluorescence intensity over time (F/Fmin). See also Supplementary Video 4. Dashed regions of interest represent fluorescent segments of extended time scales. Both control and dKO traces reveal faster oscillatory frequencies in response to exendin-4. D: The corresponding area under the curve (AUC) is also presented (AU, arbitrary units) (n = 19–20 islets, three mice per genotype). The 10G AUC was measured between 200 s and 660 s, 10G+ex4 AUC was measured between 800 s and 950 s, and KCl AUC was measured between 1,200 s and 1,500 s (AUC 10G: control vs. dKO, P = 0.09; AUC control: 10G vs. ex4, P < 0.05; AUC dKO: 10G vs. ex4, P < 0.001; ex4 vs. KCl, P < 0.05). E: Dissociated β-cells were transfected with D4ER to measure changes in [Ca2+]ER, and perifused with 10G, 10G+ex4, or thapsigargin (10G+thapsi), as indicated. Traces represent corrected ratio values postlinear fitting over time. F: AUC was measured between 350 s and 900 s (under 10G+ex4) and between 900 s and 1,300 s (10G+thapsi) from the data shown in E (n = 44–46 cells, four to five mice per genotype). G: Changes in cytoplasmic ATP-to-ADP ratio in response to 10G or 10G with 100 nmol/L ex4 was examined in whole islets. H: AUC values corresponding to G were measured between 185 s and 720 s (under 10G exposure) or between 721 s and 1,200 s (under 10G with ex4) (data points from n = 3 mice per genotype). I: Average OCR values of islets (∼10 per well) that were exposed to 3G or 10G (final concentration), 10G supplemented with ex4, FSK, oligomycin A (Oligo), FCCP, and rotenone (Rot) with antimycin A (AA) (n = 3 mice per genotype; experiment performed in duplicate). J: Insulin secretion measured during serial incubations in batches in 3G or 17G supplemented with 100 μmol/L diazoxide and 30 mmol/L KCl, (n = 3 mice per genotype in two independent experiments). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05, **P < 0.01, ****P < 0.0001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Close modal

We next explored whether the incretin-mediated improvements in insulin secretion in response to incretins were the result of altered [Ca2+]cyt dynamics. Islets from isolated dKO mice displayed a delayed increase in [Ca2+]cyt in response to 10 mmol/L glucose compared with control islets (Fig. 6C and D). Addition of exendin-4 led to the emergence of oscillatory activity in both groups, and under these conditions, differences between genotypes, as seen in Fig. 4B, were no longer evident (Fig. 6C). Measured at 10 mmol/L glucose, control and dKO islets displayed increases in ER Ca2+ in response to exendin-4 (Fig. 6E and F), while the response was exaggerated in the latter group. Neither group displayed significant changes in the ATP-to-ADP ratio in response to exendin-4 (Fig. 6G and H). Analysis of the OCR revealed no significant differences between genotypes at 10 mmol/L glucose in the presence or absence of exendin-4 or FSK (Fig. 6I).

Moreover, mitofusin deletion may lead to a partial activation of “amplification” pathways of GSIS (46) at 3 mmol/L glucose since insulin secretion was enhanced in dKO islets after depolarization of the plasma membrane with KCl in the presence of diazoxide (Fig. 6J). Conversely, no differences between islet genotypes were observed at 17 mmol/L glucose (Fig. 6J).

While glucose-induced β-cell–β-cell connectivity, as assessed by monitoring Ca2+ dynamics (Fig. 6C), was markedly impaired in dKO islets (Fig. 7A and Supplementary Fig 7), these differences were largely abolished in the presence of exendin-4 (Fig. 8B–D).

Figure 7

The GLP1-R agonist, exendin-4, improves intercellular connectivity in βMfn1/2-dKO β-cells. A: Representative Cartesian maps of control and dKO islets with color-coded lines connecting cells according to the strength of Pearson analysis (color-coded r values from 0 to 1, blue to red, respectively) under 10 mmol/L (10G), 10G with 100 nmol/L exendin-4 (10G+ex4), or 20 mmol/L KCl; scale bars: 40 μm. B: Representative heat maps depicting connectivity strength (r) of all cell pairs according to the color-coded r values from 0 to 1, blue to yellow, respectively. C: Percentage of connected cell pairs at 10G, 10G+ex4, or KCl (n = 19–20 islets, three mice per genotype). D: The r values between β-cells in response to glucose, exendin-4, or KCl (n = 3 mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05,**P < 0.01, ***P < 0.001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Figure 7

The GLP1-R agonist, exendin-4, improves intercellular connectivity in βMfn1/2-dKO β-cells. A: Representative Cartesian maps of control and dKO islets with color-coded lines connecting cells according to the strength of Pearson analysis (color-coded r values from 0 to 1, blue to red, respectively) under 10 mmol/L (10G), 10G with 100 nmol/L exendin-4 (10G+ex4), or 20 mmol/L KCl; scale bars: 40 μm. B: Representative heat maps depicting connectivity strength (r) of all cell pairs according to the color-coded r values from 0 to 1, blue to yellow, respectively. C: Percentage of connected cell pairs at 10G, 10G+ex4, or KCl (n = 19–20 islets, three mice per genotype). D: The r values between β-cells in response to glucose, exendin-4, or KCl (n = 3 mice per genotype). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05,**P < 0.01, ***P < 0.001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Close modal
Figure 8

Insulin secretion is rescued through an EPAC-dependent activation in dKO islets. A: Insulin secretion measured during serial incubations in batches in 3 mmol/L glucose (3G), 10 mmol/L glucose (10G), or 10G supplemented with 10 μmol/L H89, or 10 µmol/l FSK with 100 µmol/l IBMX or H89 (n = 3 mice per genotype, in two independent experiments). B: Insulin secretion measured during serial incubations in batches in 10G, or 10G supplemented with 6 μmol/L EPAC-activator, or EPAC-activator with 10 μmol/L H89 (n = 3 mice per genotype, in two independent experiments). C: Representative Epac1-camps FRET traces in response to 3G or 10G, or 10G supplemented with 100 nmol/L exendin-4 (10G+ex4), or 10 µmol/l FSK with 100 µmol/l IBMX in dissociated β-cells. D: Fluorescence ratio peak values corresponding to C were measured between 200 and 250s (under 10G), 620 and 720s (under 10G with ex4) or 1,110 and 1,160s (under 10G with IBMX and FSK) (n = 3–4 mice per genotype, 15–35 cells in two independent experiments). E: Representative Epac1-camps FRET traces in response to 10G, 10G+ex4, 10 µmol/L FSK (dark blue or purple traces), or 100 µmol/L IBMX (light blue or pink traces) in dissociated β-cells (n = 3 mice per genotype, 15–45 cells). F: qRT-PCR quantification of Epac, Adcy, and Prka genes expression in control and dKO islets relative to β-actin (n = 3 mice per genotype in two independent experiments). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05,**P < 0.01, ***P < 0.001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Figure 8

Insulin secretion is rescued through an EPAC-dependent activation in dKO islets. A: Insulin secretion measured during serial incubations in batches in 3 mmol/L glucose (3G), 10 mmol/L glucose (10G), or 10G supplemented with 10 μmol/L H89, or 10 µmol/l FSK with 100 µmol/l IBMX or H89 (n = 3 mice per genotype, in two independent experiments). B: Insulin secretion measured during serial incubations in batches in 10G, or 10G supplemented with 6 μmol/L EPAC-activator, or EPAC-activator with 10 μmol/L H89 (n = 3 mice per genotype, in two independent experiments). C: Representative Epac1-camps FRET traces in response to 3G or 10G, or 10G supplemented with 100 nmol/L exendin-4 (10G+ex4), or 10 µmol/l FSK with 100 µmol/l IBMX in dissociated β-cells. D: Fluorescence ratio peak values corresponding to C were measured between 200 and 250s (under 10G), 620 and 720s (under 10G with ex4) or 1,110 and 1,160s (under 10G with IBMX and FSK) (n = 3–4 mice per genotype, 15–35 cells in two independent experiments). E: Representative Epac1-camps FRET traces in response to 10G, 10G+ex4, 10 µmol/L FSK (dark blue or purple traces), or 100 µmol/L IBMX (light blue or pink traces) in dissociated β-cells (n = 3 mice per genotype, 15–45 cells). F: qRT-PCR quantification of Epac, Adcy, and Prka genes expression in control and dKO islets relative to β-actin (n = 3 mice per genotype in two independent experiments). Experiments were performed in 14-week-old male mice. Data are presented as mean ± SD. *P < 0.05,**P < 0.01, ***P < 0.001, assessed by two-way ANOVA test and the Šidák multiple comparisons test.

Close modal

Insulin Secretion Is Rescued by Incretins Through an EPAC-Dependent Activation

To explore the actions of mitochondrial disruption on incretin signaling, we next used a pharmacological approach. GSIS was more strongly enhanced in dKO versus control islets by IBMX, FSK, or the protein kinase A (PKA) inhibitor H89 alone (Fig. 8A and Supplementary Table 4). Selective activation of EPAC also tended to lead to a larger increase in insulin secretion in dKO than in control islets, and this difference became significant when PKA was inhibited with H89 (Fig. 8B).

Glucose-dependent increases in cytosolic cAMP, assessed using the Epac-camps sensor, were also markedly amplified in dKO versus control cells (Fig. 8C and D). This difference persisted in the presence of IBMX and FSK, added separately or alone (Fig. 8C and E). No changes in the expression of Epac, Adcy, or Prkar (PKA) subunits were apparent between control and dKO islets (Fig. 8F).

Defective GSIS Is Rescued by GLP-1R Agonism in Clec16a-Null Mice

To determine whether incretins may reverse defective insulin secretion in an alternative model of mitochondrial dysfunction, we examined mice lacking the mitophagy regulator Clec16a selectively in the pancreatic islet (Clec16aΔpanc) (22). GSIS was sharply inhibited in null versus Pdx1-Cre control mice, and these differences between genotype were largely corrected in by the addition of exendin-4 (Supplementary Fig. 8A). Correspondingly, whereas the difference between Clec16aΔpanc and control mice was significant for IPGTTs, there was no such (significant) difference for the OGTTs at 15 min, in line with the findings above for βMfn1/2-dKO mice (Supplementary Fig. 8AC).

Defective Secretion of a Preserved Pool of Morphologically Docked Granules in βMfn1/2-dKO Mouse β-Cells

To determine whether the markedly weaker stimulation of insulin secretion in dKO islets may reflect failed recruitment of secretory granules into a readily releasable or morphologically docked pool beneath the plasma membrane, we next deployed total internal reflection fluorescence microscopy in dissociated β-cells. By overexpressing NPY-Venus, the number of insulin granules was significantly higher in close proximity with the plasma membrane in dKO cells after treatment with 20 mmol/L KCl (Supplementary Fig. 9A and B). However, when we then used ZIMIR (30) in response to depolarization as a surrogate for insulin secretion, release events were fewer in number and smaller in dKO (Supplementary Fig. 9CE).

Altered Plasma Metabolomic and Lipidomic Profiles in βMfn1/2-dKO Mice

We applied an -omics approach to study metabolite and lipid changes in peripheral plasma samples from control and dKO mice (Supplementary Fig. 10). Of 29 metabolites, the levels of five metabolic species (shown in red) were significantly altered in βMfn1/2-dKO animals (Supplementary Fig. 10A). In the lipidomics analysis, the majority of lipid classes displayed a remarkably homogeneous downward trend in dKO samples (Supplementary Fig. 10B).

The key goal of the current study was to determine the role of mitofusins in controlling mitochondrial dynamics and hence glucose- and incretin-stimulated insulin secretion in the β-cell. Our strategy involved deleting both mitofusin isoforms since the expression of Mfn1 and Mfn2 is similar in the β-cell (47), suggestive of partial functional redundancy (48). Our measurements of Mfn1 and Mfn2 expression in mouse models of T2D nonetheless revealed changes in the expression of these genes, which may contribute to the disease.

Importantly, we show that Mfn1 and Mfn2 are critical regulators of the mitochondrial network in β-cells and consequently of insulin secretion in vitro and in vivo (see also [41]) (Supplementary Fig. 11A and B). These findings are in line with earlier studies, albeit involving the deletion of genes other than the mitofusins (1318). Additionally, we show that changes in Mfn1 and Mfn2 expression occur in models of diabetes, and hence, their forced changes, as achieved in our study, may have relevance for the pathoetiology of β-cell failure in T2D and metabolic changes consistent with insulin deficiency. These include higher levels of bile acids as previously described in rodent models of type 1 diabetes (T1D) and T2D and in humans (49,50), elevated leucine and isoleucine, as observed in human T1D (51), and an altered triglyceride profile (52). Finally, these metabolomic/lipidomic data provide further support for the expected actions of mitofusin deletion via altered β-cell function, with changes that are somewhat more in line with metabolomic changes in human T1D (and models thereof) than T2D (53). Indeed, dKO mice gain less weight than controls as they show the classic symptoms of diabetes (54,55). This is likely to be the result of metabolic dyshomeostasis in the face of lowered circulating insulin levels, leading to impaired fat storage, loss of liver and muscle glycogen, and eventually, loss of muscle mass (i.e., the cardinal symptoms of T1D and of advanced insulin-requiring T2D in humans).

Of note, none of the earlier reports investigating the effects of mitochondrial disruption in the β-cell explored the effects on incretin-stimulated secretion. Suggesting a differential effect on glucose- versus incretin-stimulated secretion we show here; firstly, that insulin secretion and glucose excursion were less markedly affected by mitofusin knockout during OGTTs, where an incretin effect is preserved (56), than during IPGTTs. Correspondingly, insulin secretion stimulated by incretins was largely preserved in dKO cells, in contrast to the ablation of glucose-stimulated secretion (Supplementary Fig. 11C and D). Strikingly, mitofusin deletion also enhanced incretin-stimulated cytosolic cAMP increases. That this effect was preserved in the face of phosphodiesterase inhibition (IBMX) and AC activation was surprising but may reflect an increase in total AC activity or distribution in dKO cells.

While PKA suppression is considered to be either neutral or inhibitory toward GSIS in wild-type β-cells (5759), our data show a rather striking increase in insulin secretion in the presence of H89 in islets from mice of either genotype. While unexpected, and in contrast with those of others that support a role for PKA downstream of cAMP in the β-cell, Bryan and colleagues (57) provide some evidence for the stimulation of GSIS by H89 under certain conditions. Nevertheless, several studies have stressed the importance of both PKA-dependent and PKA-independent effects of increased [cAMP]i on GSIS from islets (60). Thus, PKA-independent exocytosis occurs through interactions between Epac-2/cAMP- guanine-nucleotide-exchange factor II (61,62), Rab3A, and Rim2 (proteins involved in vesicle trafficking [57,58, 63] and fusion) (64). On the other hand, GLUT2, Kir6.2, and SUR1, and α-SNAP (a vesicle-associated protein) have been reported to be phosphorylated by PKA (58). Here, we show that the effect of mitofusin deletion on GSIS is preserved when PKA is inhibited by H89 and even potentiated by EPAC-activation (Supplementary Fig. 11C and D). These changes appear to be exerted at the posttranscriptional level, since we observed no changes in levels of mRNAs encoding the relevant β-cell isoforms of Epac. Whether there are changes in the level or the corresponding proteins including EPAC, their subcellular localization or interaction with upstream regulators or downstream effectors, remains to be explored. Finally, the latter findings could indicate that an intact mitochondrial reticulum restricts signaling by EPAC through a mechanism that is inhibited by PKA. Future studies, using additional or alternative PKA inhibitors (65), will be needed to explore these possibilities.

Possibly contributing to these differences in the effects on responses to glucose versus incretin, exendin-4 treatment led to greater Ca2+ accumulation in the ER in dKO cells. By enhancing Ca2+ cycling across the ER membrane, this could conceivably drive larger local increases in cytosolic Ca2+, which, in turn, may influence plasma membrane potential, trigger Ca2+ influx via VDCCs, and hence, stimulate insulin release (66).

We also demonstrate that preserved mitochondrial ultrastructure is critical for normal β-cell–β-cell connectivity, itself required for normal insulin secretion (41,67). The mechanisms underlying impaired connectivity in the absence of mitofusins are unclear but may involve altered Cx36/Gjd2 expression, phosphorylation, or activity impacting gap junctions (42).

In summary, we show that acute treatment with incretins, commonly used as treatments for T2D and obesity (56), largely reverses the deficiencies in insulin secretion that follow mitochondrial disruption. Future studies will be needed to address the relevance of these findings to human β-cells and to the action of incretins in clinical settings.

This article contains supplementary material online at https://doi.org/10.2337/figshare.19607232.

Acknowledgments. The authors thank Stephen M. Rothery, from the Facility for Imaging by Light Microscopy (FILM) at Imperial College London, for support with confocal and widefield microscopy image recording and analysis. The authors thank Professor Julia Gorelik and Sasha Judina (Imperial College) for providing the Epac1-camps sensor, and Aida Di Gregorio from the National Heart and Lung Institute (Imperial College) for genotyping the mice.

Funding. G.A.R. was supported by a Wellcome Trust Senior Investigator Award (098424AIA) and Wellcome Trust Investigator Award (212625/Z/18/Z), Medical Research Council Programme grants (MR/R022259/1, MR/J0003042/1, MR/L020149/1), an Experimental Challenge Grant (DIVA, MR/L02036X/1), a Medical Research Council grant (MR/N00275X/1), and Diabetes UK grants (BDA/11/0004210, BDA/15/0005275, BDA16/0005485). I.L. was supported by a Diabetes UKD project grant (16/0005485). This project has received funding from the European Commission Innovative Medicines Initiative 2 Joint Undertaking, under grant agreement no. 115881 (RHAPSODY). This Joint Undertaking receives support from the European Union’s Horizon 2020 Research and Innovation Programme. This work is supported by the Swiss State Secretariat for Education, Research and Innovation (SERI), under contract no. 16.0097. A.T. was supported by Medical Research Council project grant MR/R010676/1. Intravital imaging was performed using resources and/or funding provided by National Institutes of Health grants R03 DK115990 (to A.K.L.), Human Islet Research Network UC4 DK104162 (to A.K.L., RRID:SCR_014393). BJ acknowledges support from the Academy of Medical Sciences, Society for Endocrinology, The British Society for Neuroendocrinology, the European Federation for the Study of Diabetes, an Engineering and Physical Sciences Research Council capital award, and the Medical Research Council (MR/R010676/1). S.A.S. was supported by the JDRF (CDA-2016-189, SRA-2018-539, COE-2019-861), the National Institutes of Health (R01 DK108921, U01 DK127747), and the U.S. Department of Veterans Affairs (I01 BX004444).

Duality of Interest. This Joint Undertaking receives support from the European Federation of Pharmaceutical Industries and Associations. G.A.R. has received grant funding and consultancy fees from Les Laboratoires Servier and Sun Pharmaceuticals. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. E.G. performed experiments and analyzed data. E.G. supported the completion of confocal and widefield microscopy and analysis. E.G. contributed to designing the study and writing the manuscript C.M., M.M., and A.K.L. were responsible for the in vivo intravital Ca2+ imaging in mice presented in the bioRxiv paper [41]. P.C. contributed to the analysis and manipulation of the in vivo intravital Ca2+ measurements as well as the preparation and imaging of total internal reflection fluorescence samples. E.A. and L.L.N. performed the oral gavage in live animals. A.T. performed the electron microscopy sample processing and data analysis. F.Y.S.W. and Y.A. generated and performed Monte Carlo-based signal binarization. T.S. contributed to the generation of the MATLAB script used for connectivity analysis. A.W. and C.L.-Q. contributed to the metabolomics analysis. B.J. assisted with the cAMP assays. Y.X. and G.G. performed studies with the Pdx1CreER mice. N.A. assisted with Seahorse experiment protocols. C.C.-G., C.M., and M.I. were responsible for the RNA sequencing data analysis. I.L. and T.A.R. were responsible for the maintenance of mouse colonies and final approval of the version to be published. S.A.S. performed studies with Clec16a mice. T.A.R. was involved in the design of the floxed Mfn alleles. G.A.R. designed the study and wrote the manuscript with input and final approval of the version to be published from all authors. G.A.R. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented as an oral or poster presentation at the 81st Scientific Sessions of the American Diabetes Association, virtual meeting, 25–29 June 2021; the Australasian Diabetes Congress 2021, virtual event, 11–13 August 2021; the 80th Scientific Sessions of the American Diabetes Association, virtual meeting, 12–16 June 2020; Diabetes UK Professional Conference 2019, Liverpool, U.K., 6–8 March 2019; Gordon Research Conference, New London, NH, 19–22 March 2019; Rhapsody Consortium; and the 54th Annual Meeting of the European Association for the Study of Diabetes, Berlin, Germany, 1–5 October 2018. A non–peer-reviewed version of this article was published on the bioRxiv preprint server (https://doi.org/10.1101/2020.04.22.055384) on 24 April 2020.

1.
Anderson
AJ
,
Jackson
TD
,
Stroud
DA
,
Stojanovski
D
.
Mitochondria-hubs for regulating cellular biochemistry: emerging concepts and networks
.
Open Biol
2019
;
9
:
190126
2.
Rutter
GA
,
Pullen
TJ
,
Hodson
DJ
,
Martinez-Sanchez
A
.
Pancreatic β-cell identity, glucose sensing and the control of insulin secretion
.
Biochem J
2015
;
466
:
203
218
3.
Rorsman
P
,
Ashcroft
FM
.
Pancreatic β-cell electrical activity and insulin secretion: of mice and men
.
Physiol Rev
2018
;
98
:
117
214
4.
Henquin
JC
.
Triggering and amplifying pathways of regulation of insulin secretion by glucose
.
Diabetes
2000
;
49
:
1751
1760
5.
Jones
B
,
Bloom
SR
,
Buenaventura
T
,
Tomas
A
,
Rutter
GA
.
Control of insulin secretion by GLP-1
.
Peptides
2018
;
100
:
75
84
6.
Yang
D
,
Ying
J
,
Wang
X
, et al
.
Mitochondrial dynamics: a key role in neurodegeneration and a potential target for neurodegenerative disease
.
Front Neurosci
2021
;
15
:
654785
7.
Rutter
GA
,
Rizzuto
R
.
Regulation of mitochondrial metabolism by ER Ca2+ release: an intimate connection
.
Trends Biochem Sci
2000
;
25
:
215
221
8.
Westermann
B
.
Bioenergetic role of mitochondrial fusion and fission
.
Biochim Biophys Acta
2012
;
1817
:
1833
1838
9.
Ma
K
,
Chen
G
,
Li
W
,
Kepp
O
,
Zhu
Y
,
Chen
Q
.
Mitophagy, mitochondrial homeostasis, and cell fate
.
Front Cell Dev Biol
2020
;
8
:
467
10.
Filadi
R
,
Greotti
E
,
Turacchio
G
,
Luini
A
,
Pozzan
T
,
Pizzo
P
.
On the role of mitofusin 2 in endoplasmic reticulum-mitochondria tethering
.
Proc Natl Acad Sci U S A
2017
;
114
:
E2266
E2267
11.
Rovira-Llopis
S
,
Bañuls
C
,
Diaz-Morales
N
,
Hernandez-Mijares
A
,
Rocha
M
,
Victor
VM
.
Mitochondrial dynamics in type 2 diabetes: pathophysiological implications
.
Redox Biol
2017
;
11
:
637
645
12.
Serasinghe
MN
,
Chipuk
JE
.
Mitochondrial fission in human diseases
.
Handb Exp Pharmacol
2017
;
240
:
159
188
13.
Reinhardt
F
,
Schultz
J
,
Waterstradt
R
,
Baltrusch
S
.
Drp1 guarding of the mitochondrial network is important for glucose-stimulated insulin secretion in pancreatic beta cells
.
Biochem Biophys Res Commun
2016
;
474
:
646
651
14.
Hennings
TG
,
Chopra
DG
,
DeLeon
ER
, et al
.
In vivo deletion of β-cell Drp1 impairs insulin secretion without affecting islet oxygen consumption
.
Endocrinology
2018
;
159
:
3245
3256
15.
Supale
S
,
Thorel
F
,
Merkwirth
C
, et al
.
Loss of prohibitin induces mitochondrial damages altering β-cell function and survival and is responsible for gradual diabetes development
.
Diabetes
2013
;
62
:
3488
3499
16.
Stiles
L
,
Shirihai
OS
.
Mitochondrial dynamics and morphology in beta-cells
.
Best Pract Res Clin Endocrinol Metab
2012
;
26
:
725
738
17.
Zhang
Z
,
Wakabayashi
N
,
Wakabayashi
J
, et al
.
The dynamin-related GTPase Opa1 is required for glucose-stimulated ATP production in pancreatic beta cells
.
Mol Biol Cell
2011
;
22
:
2235
2245
18.
Men
X
,
Wang
H
,
Li
M
, et al
.
Dynamin-related protein 1 mediates high glucose induced pancreatic beta cell apoptosis
.
Int J Biochem Cell Biol
2009
;
41
:
879
890
19.
Del Guerra
S
,
Lupi
R
,
Marselli
L
, et al
.
Functional and molecular defects of pancreatic islets in human type 2 diabetes
.
Diabetes
2005
;
54
:
727
735
20.
Chen
H
,
McCaffery
JM
,
Chan
DC
.
Mitochondrial fusion protects against neurodegeneration in the cerebellum
.
Cell
2007
;
130
:
548
562
21.
Gu
G
,
Dubauskaite
J
,
Melton
DA
.
Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors
.
Development
2002
;
129
:
2447
2457
22.
Soleimanpour
SA
,
Gupta
A
,
Bakay
M
, et al
.
The diabetes susceptibility gene Clec16a regulates mitophagy
.
Cell
2014
;
157
:
1577
1590
23.
Ravier
MA
,
Rutter
GA
.
Isolation and culture of mouse pancreatic islets for ex vivo imaging studies with trappable or recombinant fluorescent probes
.
Methods Mol Biol
2010
;
633
:
171
184
24.
Georgiadou
E
,
Haythorne
E
,
Dickerson
MT
, et al
.
The pore-forming subunit MCU of the mitochondrial Ca2+ uniporter is required for normal glucose-stimulated insulin secretion in vitro and in vivo in mice
.
Diabetologia
2020
;
63
:
1368
1381
25.
Kolesar
JE
,
Wang
CY
,
Taguchi
YV
,
Chou
SH
,
Kaufman
BA
.
Two-dimensional intact mitochondrial DNA agarose electrophoresis reveals the structural complexity of the mammalian mitochondrial genome
.
Nucleic Acids Res
2013
;
41
:
e58
26.
Ravier
MA
,
Daro
D
,
Roma
LP
, et al
.
Mechanisms of control of the free Ca2+ concentration in the endoplasmic reticulum of mouse pancreatic β-cells: interplay with cell metabolism and [Ca2+]c and role of SERCA2b and SERCA3
.
Diabetes
2011
;
60
:
2533
2545
27.
Varadi
A
,
Rutter
GA
.
Dynamic imaging of endoplasmic reticulum Ca2+ concentration in insulin-secreting MIN6 Cells using recombinant targeted cameleons: roles of sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)-2 and ryanodine receptors
.
Diabetes
2002
;
51
(
Suppl. 1
):
S190
S201
28.
Nikolaev
VO
,
Bünemann
M
,
Hein
L
,
Hannawacker
A
,
Lohse
MJ
.
Novel single chain cAMP sensors for receptor-induced signal propagation
.
J Biol Chem
2004
;
279
:
37215
37218
29.
Wiemerslage
L
,
Lee
D
.
Quantification of mitochondrial morphology in neurites of dopaminergic neurons using multiple parameters
.
J Neurosci Methods
2016
;
262
:
56
65
30.
Li
D
,
Chen
S
,
Bellomo
EA
, et al
.
Imaging dynamic insulin release using a fluorescent zinc indicator for monitoring induced exocytotic release (ZIMIR)
.
Proc Natl Acad Sci U S A
2011
;
108
:
21063
21068
31.
Tsuboi
T
,
Rutter
GA
.
Multiple forms of “kiss-and-run” exocytosis revealed by evanescent wave microscopy
.
Curr Biol
2003
;
13
:
563
567
32.
Ahonen
L
,
Jäntti
S
,
Suvitaival
T
, et al
.
Targeted clinical metabolite profiling platform for the stratification of diabetic patients
.
Metabolites
2019
;
9
:
184
33.
Taddeo
EP
,
Alsabeeh
N
,
Baghdasarian
S
, et al
.
Mitochondrial proton leak regulated by cyclophilin D elevates insulin secretion in islets at nonstimulatory glucose levels
.
Diabetes
2020
;
69
:
131
145
34.
Brand
MD
,
Nicholls
DG
.
Assessing mitochondrial dysfunction in cells
.
Biochem J
2011
;
435
:
297
312
35.
Carrat
GR
,
Haythorne
E
,
Tomas
A
, et al
.
The type 2 diabetes gene product STARD10 is a phosphoinositide-binding protein that controls insulin secretory granule biogenesis
.
Mol Metab
2020
;
40
:
101015
36.
Akalestou
E
,
Suba
K
,
Lopez-Noriega
L
, et al
.
Intravital imaging of islet Ca2+ dynamics reveals enhanced β cell connectivity after bariatric surgery in mice
.
Nat Commun
2021
;
12
:
5165
37.
Cruciani-Guglielmacci
C
,
Bellini
L
,
Denom
J
, et al
.
Molecular phenotyping of multiple mouse strains under metabolic challenge uncovers a role for Elovl2 in glucose-induced insulin secretion
.
Mol Metab
2017
;
6
:
340
351
38.
Benjamini
Y
,
Hochberg
Y
.
Controlling the false discovery rate: a practical and powerful approach to multiple testing
.
J R Stat Soc B
1995
;
57
:
289
300
39.
Elayat
AA
,
el-Naggar
MM
,
Tahir
M
.
An immunocytochemical and morphometric study of the rat pancreatic islets
.
J Anat
1995
;
186
:
629
637
40.
Pullen
TJ
,
Huising
MO
,
Rutter
GA
.
Analysis of purified pancreatic islet beta and alpha cell transcriptomes reveals 11β-hydroxysteroid dehydrogenase (Hsd11b1) as a novel disallowed gene
.
Front Genet
2017
;
8
:
41
41.
Georgiadou
E
,
Muralidharan
C
,
Martinez
M
, et al
.
Mitofusins Mfn1 and Mfn2 are required to preserve glucose but not incretin-stimulated beta cell connectivity and insulin secretion
.
13 August 2021 [preprint]. bioRxiv:2020.04.22.055384
42.
Rutter
GA
,
Georgiadou
E
,
Martinez-Sanchez
A
,
Pullen
TJ
.
Metabolic and functional specialisations of the pancreatic beta cell: gene disallowance, mitochondrial metabolism and intercellular connectivity
.
Diabetologia
2020
;
63
:
1990
1998
43.
Johnston
NR
,
Mitchell
RK
,
Haythorne
E
, et al
.
Beta cell hubs dictate pancreatic islet responses to glucose
.
Cell Metab
2016
;
24
:
389
401
44.
Gautam
D
,
Han
SJ
,
Hamdan
FF
, et al
.
A critical role for beta cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo
.
Cell Metab
2006
;
3
:
449
461
45.
Gautam
D
,
Ruiz de Azua
I
,
Li
JH
, et al
.
Beneficial metabolic effects caused by persistent activation of beta-cell M3 muscarinic acetylcholine receptors in transgenic mice
.
Endocrinology
2010
;
151
:
5185
5194
46.
Gembal
M
,
Gilon
P
,
Henquin
JC
.
Evidence that glucose can control insulin release independently from its action on ATP-sensitive K+ channels in mouse B cells
.
J Clin Invest
1992
;
89
:
1288
1295
47.
Benner
C
,
van der Meulen
T
,
Cacéres
E
,
Tigyi
K
,
Donaldson
CJ
,
Huising
MO
.
The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression
.
BMC Genomics
2014
;
15
:
620
48.
Sidarala
V
,
Zhu
J
,
Pearson
GL
,
Reck
EC
,
Kaufman
BA
,
Soleimanpour
SA
.
Mitofusins 1 and 2 collaborate to fuel pancreatic beta cell insulin release via regulation of both mitochondrial structure and DNA content
.
11 January 2021 [preprint]. bioRxiv 2021.01.10.426151
.
49.
Andersén
E
,
Karlaganis
G
,
Sjövall
J
.
Altered bile acid profiles in duodenal bile and urine in diabetic subjects
.
Eur J Clin Invest
1988
;
18
:
166
172
50.
Uchida
K
,
Makino
S
,
Akiyoshi
T
.
Altered bile acid metabolism in nonobese, spontaneously diabetic (NOD) mice
.
Diabetes
1985
;
34
:
79
83
51.
Sailer
M
,
Dahlhoff
C
,
Giesbertz
P
, et al
.
Increased plasma citrulline in mice marks diet-induced obesity and may predict the development of the metabolic syndrome
.
PLoS One
2013
;
8
:
e63950
52.
Lamichhane
S
,
Ahonen
L
,
Dyrlund
TS
, et al
.
Dynamics of plasma lipidome in progression to islet autoimmunity and type 1 diabetes - Type 1 Diabetes Prediction and Prevention Study (DIPP)
.
Sci Rep
2018
;
8
:
10635
53.
George
AM
,
Jacob
AG
,
Fogelfeld
L
.
Lean diabetes mellitus: an emerging entity in the era of obesity
.
World J Diabetes
2015
;
6
:
613
620
54.
Mitchell
RK
,
Nguyen-Tu
MS
,
Chabosseau
P
, et al
.
The transcription factor Pax6 is required for pancreatic β cell identity, glucose-regulated ATP synthesis, and Ca2+ dynamics in adult mice
.
J Biol Chem
2017
;
292
:
8892
8906
55.
Martinez-Sanchez
A
,
Nguyen-Tu
M-S
,
Rutter
GA
.
DICER Inactivation Identifies Pancreatic β-Cell “Disallowed” Genes Targeted by MicroRNAs
.
Mol Endocrinol
2015
;
29
:
1067
1079
56.
Nauck
MA
,
Quast
DR
,
Wefers
J
,
Meier
JJ
.
GLP-1 receptor agonists in the treatment of type 2 diabetes - state-of-the-art
.
Mol Metab
2021
;
46
:
101102
57.
Nakazaki
M
,
Crane
A
,
Hu
M
, et al
.
cAMP-activated protein kinase-independent potentiation of insulin secretion by cAMP is impaired in SUR1 null islets
.
Diabetes
2002
;
51
:
3440
3449
58.
Kashima
Y
,
Miki
T
,
Shibasaki
T
, et al
.
Critical role of cAMP-GEFII--Rim2 complex in incretin-potentiated insulin secretion
.
J Biol Chem
2001
;
276
:
46046
46053
59.
Chepurny
OG
,
Kelley
GG
,
Dzhura
I
, et al
.
PKA-dependent potentiation of glucose-stimulated insulin secretion by Epac activator 8-pCPT-2′-O-Me-cAMP-AM in human islets of Langerhans
.
Am J Physiol Endocrinol Metab
2010
;
298
:
E622
E633
60.
Renström
E
,
Eliasson
L
,
Rorsman
P
.
Protein kinase A-dependent and -independent stimulation of exocytosis by cAMP in mouse pancreatic B-cells
.
J Physiol
1997
;
502
:
105
118
61.
de Rooij
J
,
Zwartkruis
FJ
,
Verheijen
MH
, et al
.
Epac is a Rap1 guanine-nucleotide-exchange factor directly activated by cyclic AMP
.
Nature
1998
;
396
:
474
477
62.
Kawasaki
H
,
Springett
GM
,
Mochizuki
N
, et al
.
A family of cAMP-binding proteins that directly activate Rap1
.
Science
1998
;
282
:
2275
2279
63.
Ozaki
N
,
Shibasaki
T
,
Kashima
Y
, et al
.
cAMP-GEFII is a direct target of cAMP in regulated exocytosis
.
Nat Cell Biol
2000
;
2
:
805
811
64.
Wang
Y
,
Perfetti
R
,
Greig
NH
, et al
.
Glucagon-like peptide-1 can reverse the age-related decline in glucose tolerance in rats
.
J Clin Invest
1997
;
99
:
2883
2889
65.
Lochner
A
,
Moolman
JA
.
The many faces of H89: a review
.
Cardiovasc Drug Rev
2006
;
24
:
261
274
66.
Gilon
P
,
Arredouani
A
,
Gailly
P
,
Gromada
J
,
Henquin
JC
.
Uptake and release of Ca2+ by the endoplasmic reticulum contribute to the oscillations of the cytosolic Ca2+ concentration triggered by Ca2+ influx in the electrically excitable pancreatic B-cell
.
J Biol Chem
1999
;
274
:
20197
20205
67.
Salem
V
,
Silva
LD
,
Suba
K
, et al
.
Leader β-cells coordinate Ca2+ dynamics across pancreatic islets in vivo
.
Nat Metab
2019
;
1
:
615
629
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.