Throughout evolution, proinsulin has exhibited significant sequence variation in both C-peptide and insulin moieties. As the proinsulin coding sequence evolves, the gene product continues to be under selection pressure both for ultimate insulin bioactivity and for the ability of proinsulin to be folded for export through the secretory pathway of pancreatic β-cells. The substitution proinsulin-R(B22)E is known to yield a bioactive insulin, although R(B22)Q has been reported as a mutation that falls within the spectrum of mutant INS-gene–induced diabetes of youth. Here, we have studied mice expressing heterozygous (or homozygous) proinsulin-R(B22)E knocked into the Ins2 locus. Neither females nor males bearing the heterozygous mutation developed diabetes at any age examined, but subtle evidence of increased proinsulin misfolding in the endoplasmic reticulum is demonstrable in isolated islets from the heterozygotes. Moreover, males have indications of glucose intolerance, and within a few weeks of exposure to a high-fat diet, they developed frank diabetes. Diabetes was more severe in homozygotes, and the development of disease paralleled a progressive heterogeneity of β-cells with increasing fractions of proinsulin-rich/insulin-poor cells as well as glucagon-positive cells. Evidently, subthreshold predisposition to proinsulin misfolding can go undetected but provides genetic susceptibility to diet-induced β-cell failure.

Several groups have been pursuing the molecular mechanisms underlying β-cell dysfunction and compensatory cellular responses in the rare genetic syndrome mutant INS-gene–induced diabetes of youth (MIDY) (13). The fundamental defect in MIDY is observed in humans (4), large animal models (5), and small animal models (6) and has been replicated in cell culture (7) in vitro (8) and modeled in silico (9). The clinical problem originates from the fact that misfolded proinsulin within the endoplasmic reticulum (ER) can propagate its misfolding and ER retention onto wild-type (WT) bystander proinsulin molecules, thereby impairing insulin production (10,11). Yet, MIDY is a rare disease (12,13).

Proinsulin exhibits significant sequence variation throughout evolution, including within both the C-peptide and the insulin moieties (14,15). The Ins gene product continues to be under selection pressure both for the ultimate bioactivity (of insulin) and for the ability of proinsulin to be folded for export through the secretory pathway of pancreatic β-cells (16). Evidence suggests that with only natural variation provided by evolution, “WT” proinsulin itself is capable of forming nonnative disulfide-linked proinsulin complexes not unlike those triggered by MIDY proinsulin mutations (17). However, massive formation of disulfide-linked complexes of WT proinsulin has thus far been observed only in the islets of db/db or other leptin receptor–deficient mice (or in the islets of normal animals that have been treated with one or more toxins that drastically perturb ER homeostasis [17]). We have also wondered whether it is possible that subtle predisposition to proinsulin misfolding can go unnoticed and yet be a genetic risk to diet-induced diabetes.

With these considerations in mind, we have interest in proinsulin residue 46, i.e., Arg at position 22 of the insulin B chain. Recent studies indicated that insulin-R(B22)Q is a naturally occurring variant in bats (15), has greater than or equal to half of the normal affinity for insulin receptor binding, and can be found released into the bloodstream of patients who express the INS c.137G>A (R46Q) variant (18). Yet in all three family members bearing this heterozygous mutation, diabetes was ultimately diagnosed at ages 17–20 years (19), suggesting that this substitution, albeit less severe than MIDY mutants triggering neonatal diabetes (12), still trips over the diabetogenic threshold.

It has been reported that modification of R(B22) with a bulky group of the opposite charge does not alter the specific bioactivity of insulin (20) as in the case of R(B22)D substitution (21), which is naturally occurring in mole rats and guinea pigs (15). Similarly, the R(B22)E substitution has also been reported to retain insulin bioactivity (22). Here, we pursued the possible impact of proinsulin-R(B22)E expression in the β-cells of mice in which this variant is knocked into the Ins2 locus. As expected, the substitution creates a proinsulin that is secretable from pancreatic β-cells, and under normal laboratory conditions, all heterozygous animals remain diabetes free. Nevertheless, heterozygous males consistently developed diabetes upon exposure to a high-fat diet (HFD), highlighting the impact of a subthreshold genetic predisposition to proinsulin misfolding on the development of diet-induced diabetes.

Proinsulin Mutagenesis

Plasmids encoding untagged human proinsulin-R(B22)E, untagged human proinsulin-WT, or Myc-tagged proinsulins were generated as previously described (23,24). All proinsulin-expressing constructs were confirmed by direct DNA sequencing.

Cell Transfection, Metabolic Labeling, Immunoprecipitation, Coimmunoprecipitation, Western Blotting

Min6 mouse β-cells (25) (obtained from Dr. D. Stoffers, University of Pennsylvania, Philadelphia, PA) were cultured in DMEM supplemented with 10% FBS, penicillin/streptomycin, and 0.05 mmol/L β-mercaptoethanol. Cells at 70–80% confluency were transfected using Lipofectamine 2000 (Thermo Fisher Scientific), with fresh media changed at 6 h posttransfection. Media were removed at 48 h; the cells were washed with ice-cold PBS and lysed in radioimmunoprecipitation assay buffer (10 mmol/L Tris, pH 7.4, 150 mmol/L NaCl, 0.1% SDS, 1% NP40, 2 mmol/L EDTA) plus protease inhibitor/phosphatase inhibitor cocktail (Sigma-Aldrich). Total protein was measured by bicinchoninic acid assay. Proteins in sample buffer were boiled and resolved by 4–12% NuPAGE Bis-Tris Gel (Invitrogen) and electrotransferred to nitrocellulose (Bio-Rad). Membranes were incubated with primary antibodies (4°C overnight) and then secondary peroxidase-conjugated goat anti-rabbit IgG (111-035-144; Jackson ImmunoResearch) or peroxidase-conjugated goat anti-mouse IgG (115-035-174; Jackson ImmunoResearch) followed by enhanced chemiluminescence (SuperSignal West Pico PLUS; Thermo Fisher Scientific) with digital image capture.

Construction of Knockin Mouse

Initially, the Mouse Genetics Core (National Jewish Health) prepared eggs from superovulated NOD females fertilized with male NOD sperm in vitro. After overnight culture, fertilized embryos were injected with a mixture of 1) Cas9 protein, 2) a CRISPR guide RNA (designed using CRISPOR software; http://crispor.tefor.net) matching a region just upstream of the proinsulin R(B22) codon, and 3) an Ins2-specific 150-base pair repair oligonucleotide covering this region, replacing the R codon (CGT) with E (GAG), plus a silent mutation, creating an Alu I restriction site. Injected embryos were introduced into pseudopregnant mice; DNA from the pups were analyzed by PCR, restriction digest, and sequencing to distinguish specific Ins2 (or offsite Ins1) locus repair with the homologous sequence versus nonhomologous end-joining events. To isolate the proper genetic event, breeding resulted in NOD mice bearing a single heterozygous Ins2 R(B22)E mutation (WT at three other Ins alleles). Breeding was then initiated to move the mutation into the C57BL6/J background. Animals were phenotyped in each of the first five generations of C57BL6/J backcrosses. All data in this article come after five generations of backcrossing (and further backcrosses are ongoing), but because the same phenotype was observed in each backcross generation, the data are reported here. HFD (5.5–11.5 weeks of age) was irradiated rodent chow (60 kcal% fat, #D12492; Research Diets, New Brunswick, NJ).

Circulating Insulin and Proinsulin, In Vivo Glucose Tolerance, and Glucose-Stimulated Insulin Secretion

ELISA was used to measure mouse insulin (80-INSMS-E10; ALPCO) and proinsulin (80-PINMS-E01; ALPCO). For in vivo glucose tolerance, mice were fasted for 6 h; glucose (1 g/kg body weight) was administered intraperitoneally; and tail vein glucose was monitored (One-Touch Ultra blood glucose meter and test strips). For in vivo glucose-stimulated insulin secretion (GSIS), serum was collected under basal conditions and under glucose-stimulated conditions at t = 15 min.

Islet Isolation and GSIS

Islets were isolated by collagenase digestion through the common bile duct followed by pancreatic digestion ex vivo. The digest was washed and spun on a Histopaque 1077 (Sigma-Aldrich) gradient (900g × 20 min without brake). Islets were collected, washed, handpicked, and incubated overnight in RPMI medium plus 10% FBS at 37°C. Recovered islets were preincubated at 2.8 mmol/L glucose for 1 h at 37°C in modified Krebs-Ringer bicarbonate buffer plus 20 mmol/L HEPES (KRBH) and 0.05% BSA. Fifteen to 17 islets were transferred to microfuge tubes containing 500 μL of KRBH-BSA solution and incubated at 37°C for 30 min at 2.8 mmol/L glucose (basal) followed by 30 min at 16.7 mmol/L glucose (stimulated), with media measured for insulin content.

Metabolic Labeling of Mouse Pancreatic Islets

Twenty-five islets isolated from WT and proinsulin-R(B22)E heterozygous and homozygous littermates were washed in prewarmed Met/Cys-deficient RPMI medium and then pulse labeled with 35S-amino acids (Tran35S label) for 30 min at 37°C. Labeled islets were either lysed immediately or chased in complete growth media for 2 h. Islets were sonicated in radioimmunoprecipitation assay buffer (25 mmol/L Tris, pH 7.5, 100 nmol/L NaCl, 1% Triton X-100, 0.2% deoxycholic acid, 0.1% SDS, 10 mmol/L EDTA) containing 2 mmol/L N-ethylmaleimide and a protease inhibitor cocktail. Lysates were normalized to trichloroacetic acid–precipitable counts and immunoprecipitated with guinea pig polyclonal anti-insulin and protein A agarose overnight at 4°C. Immunoprecipitates were washed and analyzed by nonreducing/reducing Tris-tricine-urea-SDS-PAGE, followed by phosphorimaging, and bands were quantified with ImageJ software.

Immunofluorescence

Paraffin sections of formaldehyde-fixed pancreas were deparaffinized with CitriSolv (Thermo Fisher Scientific) and rehydrated in a decreasing graded series of ethanol followed by heating for antigen retrieval. Slides were washed with PBS and incubated in blocking buffer (Tris-buffered saline [TBS] plus 0.2% Triton X-100 and 3% BSA) for 2 h and incubated in primary antibody (in TBS plus 3% BSA and 0.2% Tween 20) overnight at 4°C. After washes, secondary antibody was incubated for 1 h at room temperature. Slides were washed three times with TBS/0.1% Tween 20, mounted with ProLong Gold plus DAPI, and imaged by epifluorescence on a Nikon A1 confocal microscope with a 60× oil objective.

Electron Microscopy

Isolated islets were fixed with 2.5% (v/v) glutaraldehyde in 0.1 mol/L sodium cacodylate buffer (CB) (pH 7.2), embedded in 2.5% low-melting agarose, trimmed to ∼1-mm cubes, washed three times in CB, and postfixed in 2% osmium tetroxide plus 1.5% potassium ferrocyanide in 0.1 mol/L CB for 1 h on ice. After three further washes in CB, cubes were washed three times in 0.1 mol/L sodium acetate buffer (pH 5.2) and then stained in this buffer containing 2% uranyl acetate for 1 h. With agitation, the cubes were washed extensively and then dehydrated in a graded series of ethanol up to 100% and then acetone for 15 min before infiltration with graded concentrations of Spurr’s resin in acetone over 3 days. Islets were finally embedded in fresh Spurr’s resin and polymerized for 45 h at 70–75°C. Ultrathin (70-nm) sections were captured on carbon-coated 200-mesh copper grids, and imaged on a JEM-1400plus transmission electron microscope (JEOL USA Inc., Peabody, MA) at 80 kV, with images captured on an AMT XR401 camera (Advanced Microscopy Techniques, Woburn, MA).

Statistical Analyses

Statistical analyses were carried out by two-tailed Student t test or one-way ANOVA followed by multiple comparison testing (GraphPad Prism 8 software). P < 0.05 was taken as statistically significant.

Data and Resource Availability

Key resources are shown in Table 1. Data generated and analyzed during the current study are contained within the figures. Additional data are available from the corresponding author upon request.

Table 1

Key resources

Reagent type (species) or resourceDesignationSource or referenceIdentifierAdditional information
Cell line (mouse) Min6 (mouse β-cell line) Soleimanpour et al.56    
Antibody Anti-rat proinsulin monoclonal CCI-17 ALPCO CCI-17 1:1,000 
Antibody Anti-human proinsulin Abmart (Haataja et al.34 1B24, 3L10 1:1,000 
Antibody Guinea pig anti-insulin Covance  1:500 
Antibody Mouse antiglucagon Abcam ab109888 1:1,000 
Antibody Antiglucagon (rabbit polyclonal) Millipore AB932 1:500 
Antibody Anti-KDEL (rabbit monoclonal) Novus Biologicals NBP2–75549 1:300 
Antibody Anti-Myc (rabbit polyclonal) Immunology Consultants Laboratory RMYC-45A 1:500 
Antibody Rabbit anti-cyclophilin B Thermo Fisher Scientific PA1-027A 1:1,000 
Antibody Alexa Fluor 488, 555, 647 (secondary) Invitrogen  1:500 
Antibody HRP guinea pig, rabbit, or mouse 2° Jackson ImmunoResearch  1:5,000 
Recombinant DNA reagent pTARGET (vector) Promega A1410  
Recombinant DNA reagent Myc-WT proinsulin (plasmid) Liu et al.57   Human proinsulin 
Commercial assay or kit QuikChange II Site-Directed Mutagenesis Kit Agilent Technologies Agilent: 200524  
Commercial assay or kit Mouse insulin ELISA ALPCO 80-INSMS-E10  
Commercial assay or kit Enhanced chemiluminescence Immobilon or Clarity Millipore or Bio-Rad, respectively   
Chemical compound, drug N-ethylmaleimide Sigma-Aldrich E3876  
Chemical compound, drug Trans 35S Label Perkin-Elmer PerkinElmer:
NEG072007MC 
 
Chemical compound, drug ProSieve 50 Gel Solution Lonza Lonza: 50618  
Chemical compound, drug cOmplete Mini Protease Inhibitor Cocktail Roche Roche:
11836153001 
 
Chemical compound, drug Collagenase P Sigma-Aldrich 11249002001  
Chemical compound, drug Protein A agarose Invitrogen   
Chemical compound, drug Dithiothreitol, pansorbin, urea Sigma-Aldrich or Thermo Fisher Scientific   
Reagent Lipofectamine 2000 Thermo Fisher Scientific   
Reagent Tissue culture reagents Thermo Fisher Scientific   
Reagent Met/Cys-deficient RPMI medium Sigma-Aldrich   
Reagent ProLong Gold Antifade Reagent with DAPI Thermo Fisher Scientific Invitrogen P36931  
Reagent Unmasking solution Thermo Fisher Scientific Cell Signaling 14747P  
Cell culture imaging Nunc LabTek-II Chambers Thermo Fisher Scientific   
Software, algorithm GraphPad Prism 8.0 GraphPad Software   
Reagent type (species) or resourceDesignationSource or referenceIdentifierAdditional information
Cell line (mouse) Min6 (mouse β-cell line) Soleimanpour et al.56    
Antibody Anti-rat proinsulin monoclonal CCI-17 ALPCO CCI-17 1:1,000 
Antibody Anti-human proinsulin Abmart (Haataja et al.34 1B24, 3L10 1:1,000 
Antibody Guinea pig anti-insulin Covance  1:500 
Antibody Mouse antiglucagon Abcam ab109888 1:1,000 
Antibody Antiglucagon (rabbit polyclonal) Millipore AB932 1:500 
Antibody Anti-KDEL (rabbit monoclonal) Novus Biologicals NBP2–75549 1:300 
Antibody Anti-Myc (rabbit polyclonal) Immunology Consultants Laboratory RMYC-45A 1:500 
Antibody Rabbit anti-cyclophilin B Thermo Fisher Scientific PA1-027A 1:1,000 
Antibody Alexa Fluor 488, 555, 647 (secondary) Invitrogen  1:500 
Antibody HRP guinea pig, rabbit, or mouse 2° Jackson ImmunoResearch  1:5,000 
Recombinant DNA reagent pTARGET (vector) Promega A1410  
Recombinant DNA reagent Myc-WT proinsulin (plasmid) Liu et al.57   Human proinsulin 
Commercial assay or kit QuikChange II Site-Directed Mutagenesis Kit Agilent Technologies Agilent: 200524  
Commercial assay or kit Mouse insulin ELISA ALPCO 80-INSMS-E10  
Commercial assay or kit Enhanced chemiluminescence Immobilon or Clarity Millipore or Bio-Rad, respectively   
Chemical compound, drug N-ethylmaleimide Sigma-Aldrich E3876  
Chemical compound, drug Trans 35S Label Perkin-Elmer PerkinElmer:
NEG072007MC 
 
Chemical compound, drug ProSieve 50 Gel Solution Lonza Lonza: 50618  
Chemical compound, drug cOmplete Mini Protease Inhibitor Cocktail Roche Roche:
11836153001 
 
Chemical compound, drug Collagenase P Sigma-Aldrich 11249002001  
Chemical compound, drug Protein A agarose Invitrogen   
Chemical compound, drug Dithiothreitol, pansorbin, urea Sigma-Aldrich or Thermo Fisher Scientific   
Reagent Lipofectamine 2000 Thermo Fisher Scientific   
Reagent Tissue culture reagents Thermo Fisher Scientific   
Reagent Met/Cys-deficient RPMI medium Sigma-Aldrich   
Reagent ProLong Gold Antifade Reagent with DAPI Thermo Fisher Scientific Invitrogen P36931  
Reagent Unmasking solution Thermo Fisher Scientific Cell Signaling 14747P  
Cell culture imaging Nunc LabTek-II Chambers Thermo Fisher Scientific   
Software, algorithm GraphPad Prism 8.0 GraphPad Software   

Expression of Recombinant Proinsulin-R(B22)E

Proinsulin-R(B22)E (Fig. 1A) should lead to bioactive insulin (2022), yet it may not necessarily support efficient proinsulin protein folding (16,2629). We first examined the behavior of recombinant human proinsulin-R(B22)E. Min6 pancreatic β-cells express their own endogenous mouse proinsulin, but we probed transfected cells, and media, with an antibody that recognizes only human proinsulin protein. Thus, from Min6 cells transfected with empty vector, no proinsulin band was detected (Fig. 1B). Both human WT and R(B22)E proinsulins were secreted from Min6 cells (Fig. 1B), and under these conditions, proinsulin-R(B22)E did not block WT proinsulin secretion (Fig. 1C). Nevertheless, from Western blotting of reducing SDS-PAGE, it was clear that the secretion efficiency (extracellular-to-intracellular ratio) for WT proinsulin was greater than that for proinsulin-R(B22)E, whereas Akita mutant proinsulin was not secreted at all (Fig. 1B and C). Thus, in Min6 β-cells, proinsulin-R(B22)E passes ER quality control, and the dominant-negative phenotype of MIDY (1) is inapparent.

Figure 1

Insulin B-chain substitution R(B22)E. A: Schematic representation of insulin chains showing the respective disulfide bonds and the R(B22)E substitution. B: Transfection of Min6 cells with empty vector (EV), untagged WT human proinsulin, hPro-R(B22)E-CpepMyc, or hPro-C(A7)Y-CpepMyc (Akita-Myc). The media-bathing transfected cells were collected overnight, and both cell lysates and media were resolved by SDS-PAGE under reducing conditions and immunoblotting with anti-human-proinsulin or cyclophilin B (CypB) (loading control) (bottom panels). Cell lysates were similarly analyzed under nonreducing conditions to reveal disulfide-linked proinsulin complexes (top panel). C: Transfection of Min6 cells as in B (first four lanes) or cotransfection with both untagged WT human proinsulin and hPro-R(B22)E-CpepMyc (last lane). Proinsulin-R(B22)E is secreted from Min6 cells, and under these conditions, proinsulin-R(B22)E does not block the secretion of coexpressed untagged WT proinsulin.

Figure 1

Insulin B-chain substitution R(B22)E. A: Schematic representation of insulin chains showing the respective disulfide bonds and the R(B22)E substitution. B: Transfection of Min6 cells with empty vector (EV), untagged WT human proinsulin, hPro-R(B22)E-CpepMyc, or hPro-C(A7)Y-CpepMyc (Akita-Myc). The media-bathing transfected cells were collected overnight, and both cell lysates and media were resolved by SDS-PAGE under reducing conditions and immunoblotting with anti-human-proinsulin or cyclophilin B (CypB) (loading control) (bottom panels). Cell lysates were similarly analyzed under nonreducing conditions to reveal disulfide-linked proinsulin complexes (top panel). C: Transfection of Min6 cells as in B (first four lanes) or cotransfection with both untagged WT human proinsulin and hPro-R(B22)E-CpepMyc (last lane). Proinsulin-R(B22)E is secreted from Min6 cells, and under these conditions, proinsulin-R(B22)E does not block the secretion of coexpressed untagged WT proinsulin.

Close modal

Endogenous Islet Expression of Ins2-Proinsulin-R(B22)E

A CRISPR/Cas9-mediated knockin of Ins2-proinsulin-R(B22)E was back-bred to C57BL6/J mice. From 5.5 to 11.5 weeks of age, WT and heterozygous Ins2-proinsulin-R(B22)E mice gained weight on a normal chow diet and as expected, gained more weight on an HFD, without statistical differences seen between the genotypes (Fig. 2A). Glucose tolerance in heterozygous Ins2-proinsulin-R(B22)E mice at 5.5 weeks of age was normal (Fig. 2B) with a normal area under the curve (Fig. 2C). However, within 2 weeks of HFD, male heterozygotes had an average random blood glucose ≥350 mg/dL, suggesting onset of diabetes (Fig. 2D). At 11.5 weeks of age, male heterozygotes on HFD for 6 weeks had clearly diagnosed diabetes (either in terms of fasting hyperglycemia, 2-h glucose tolerance, or area under the curve), whereas those on a normal chow diet did not meet any diabetes criteria (Fig. 2E and F). Despite that random glucose in HFD-fed male heterozygotes was higher, serum insulin was not elevated (Fig. 2G); actually, the random serum insulin-to-glucose ratio was significantly lower in HFD-fed male heterozygotes (Fig. 2H). Indeed, after 6 weeks of HFD, isolated islets from male Ins2-proinsulin-R(B22)E heterozygotes exhibited abnormally low insulin secretion (unstimulated and glucose-stimulated) despite a normal fold change (Fig. 2I). Islets from male heterozygotes also developed diminished insulin content, and in HFD-fed animals, insulin content fell farther (Fig. 2J). Interestingly, proinsulin in these mice (Fig. 2J) was preserved, although proinsulin formed enhanced nonnative disulfide-linked complexes (Supplementary Fig. 1A and B). In contrast, female Ins2-proinsulin-R(B22)E heterozygotes gained weight normally and did not develop random hyperglycemia on HFD (Fig. 2K and L).

Figure 2

Heterozygous (Het) Ins2-proinsulin-R(B22)E mice fed normal chow (NC) or HFD. A: Weekly body weight measurements in male mice (n = 4–8 per group). B: Intraperitoneal glucose tolerance test in 5.5-week-old males on NC (n = 4–5 per group). C: Area under the curve (AUC) from the data in B. D: Weekly random blood glucose in males on NC or HFD (n = 4–8 per group), with Ins2-proinsulin-R(B22)E-Het on NC shown in blue and on HFD in red. E: Intraperitoneal glucose tolerance test in 11-week-old males on NC or HFD for 6 weeks (n = 3–5 per group), with Ins2-proinsulin-R(B22)E-Het mice on NC in blue and on HFD in red. F: AUC from the data in E. G: Random serum insulin level in 11-week-old WT (purple) or Ins2-proinsulin-R(B22)E-Het (red) males on HFD for 6 weeks (n = 5–7 per group). H: The data from G used to calculate serum insulin-to-glucose ratio. I: Insulin secretion from isolated islets of WT (purple) or Ins2-proinsulin-R(B22)E-Het (red) males on HFD at 2.8 mmol/L (unstimulated) and 16.7 mmol/L (stimulated) glucose (n = 3–4 per group). J: Isolated islets from male WT and Ins2-proinsulin-R(B22)E-Het mice fed NC or HFD were lysed and analyzed by reducing SDS-PAGE and immunoblotting with monoclonal antibody anti-proinsulin (top panel), guinea pig anti-insulin (middle panel), and anti-cyclophilin B (CypB) (loading control) (bottom panel). K: Weekly body weight measurements in female Ins2-proinsulin-R(B22)E-Het mice fed NC (blue) or HFD (green) (n = 4 per group). L: Weekly random blood glucose measurements in female Ins2-proinsulin-R(B22)E-Het mice on NC (blue) or on HFD (green) (n = 4 per group). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 by one-way ANOVA.

Figure 2

Heterozygous (Het) Ins2-proinsulin-R(B22)E mice fed normal chow (NC) or HFD. A: Weekly body weight measurements in male mice (n = 4–8 per group). B: Intraperitoneal glucose tolerance test in 5.5-week-old males on NC (n = 4–5 per group). C: Area under the curve (AUC) from the data in B. D: Weekly random blood glucose in males on NC or HFD (n = 4–8 per group), with Ins2-proinsulin-R(B22)E-Het on NC shown in blue and on HFD in red. E: Intraperitoneal glucose tolerance test in 11-week-old males on NC or HFD for 6 weeks (n = 3–5 per group), with Ins2-proinsulin-R(B22)E-Het mice on NC in blue and on HFD in red. F: AUC from the data in E. G: Random serum insulin level in 11-week-old WT (purple) or Ins2-proinsulin-R(B22)E-Het (red) males on HFD for 6 weeks (n = 5–7 per group). H: The data from G used to calculate serum insulin-to-glucose ratio. I: Insulin secretion from isolated islets of WT (purple) or Ins2-proinsulin-R(B22)E-Het (red) males on HFD at 2.8 mmol/L (unstimulated) and 16.7 mmol/L (stimulated) glucose (n = 3–4 per group). J: Isolated islets from male WT and Ins2-proinsulin-R(B22)E-Het mice fed NC or HFD were lysed and analyzed by reducing SDS-PAGE and immunoblotting with monoclonal antibody anti-proinsulin (top panel), guinea pig anti-insulin (middle panel), and anti-cyclophilin B (CypB) (loading control) (bottom panel). K: Weekly body weight measurements in female Ins2-proinsulin-R(B22)E-Het mice fed NC (blue) or HFD (green) (n = 4 per group). L: Weekly random blood glucose measurements in female Ins2-proinsulin-R(B22)E-Het mice on NC (blue) or on HFD (green) (n = 4 per group). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 by one-way ANOVA.

Close modal

When Ins2-proinsulin-R(B22)E was bred to homozygosity (still leaving both WT Ins1 alleles), both males and females at 5 weeks of age exhibited random blood glucose that averaged in the diabetic range, even on a normal chow diet (Fig. 3A), and fasting blood glucose values were similarly elevated. (Random blood glucose in homozygotes rose even higher by 8 weeks of age, whereas heterozygotes remained normoglycemic [Fig. 3B].) Despite the higher blood glucose, neither homozygous males nor females had raised endogenous serum insulin; with both sexes combined, it was apparent that homozygotes had decreased insulinemia despite ongoing hyperglycemic stimulation (Fig. 3C), with a profound lowering of serum insulin-to-glucose ratio (Fig. 3D). These three genotypes were clearly separable when comparing acute GSIS in vivo (Fig. 3E). Moreover, isolated islets from these subgroups tested for GSIS in vitro were distinct (Fig. 3F), despite that the fold change of GSIS was not statistically different. The data indicate a decline of releasable insulin beginning with Ins2-proinsulin-R(B22)E heterozygotes and worsening in homozygotes.

Figure 3

Homozygous (Hom) Ins2-proinsulin-R(B22)E mice develop spontaneous diabetes. A: Random blood glucose at 5 weeks of age in WT (black), heterozygous (Het) (blue), and Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow (n = 3–7 per group). Males and females shown separately as well as combined. B: The same mice from A remeasured 8 weeks postpartum. C: Serum insulin level measured at 5.5 weeks of age in WT (black), Het (blue), or Hom (red) males fed normal chow (n = 5 per group). Males and females shown separately as well as combined. D: Insulin-to-glucose ratio calculated from the samples in B and C, respectively. E: GSIS in vivo at 0 and 15 min poststimulation from WT (black), Het (blue), or Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow (6-week-old males, n = 5–6 per group). F: GSIS from islets isolated from WT (black), Het (blue), or Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow at 2.8 mmol/L (unstimulated) and 16.7 mmol/L (stimulated) glucose concentration (6-week-old males, n = 5–7 per group). G: Serum proinsulin level from the same animals measured in C (males and females combined). H: Proinsulin-to-insulin ratio from the animals in G (males and females combined). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 by one-way ANOVA.

Figure 3

Homozygous (Hom) Ins2-proinsulin-R(B22)E mice develop spontaneous diabetes. A: Random blood glucose at 5 weeks of age in WT (black), heterozygous (Het) (blue), and Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow (n = 3–7 per group). Males and females shown separately as well as combined. B: The same mice from A remeasured 8 weeks postpartum. C: Serum insulin level measured at 5.5 weeks of age in WT (black), Het (blue), or Hom (red) males fed normal chow (n = 5 per group). Males and females shown separately as well as combined. D: Insulin-to-glucose ratio calculated from the samples in B and C, respectively. E: GSIS in vivo at 0 and 15 min poststimulation from WT (black), Het (blue), or Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow (6-week-old males, n = 5–6 per group). F: GSIS from islets isolated from WT (black), Het (blue), or Hom (red) Ins2-proinsulin-R(B22)E mice fed normal chow at 2.8 mmol/L (unstimulated) and 16.7 mmol/L (stimulated) glucose concentration (6-week-old males, n = 5–7 per group). G: Serum proinsulin level from the same animals measured in C (males and females combined). H: Proinsulin-to-insulin ratio from the animals in G (males and females combined). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 by one-way ANOVA.

Close modal

Porte and Kahn (30) as well as others (31) reported that patients with type 2 diabetes (T2D) demonstrate disproportionately elevated levels of circulating proinsulin that tends to be worse with the degree of hyperglycemia and correlates with a diminished maximal insulin secretion capacity. Yet, recent reports have suggested that serum proinsulin-to-insulin ratio measurements may be of limited value across populations (32,33), although they may still be of value within selected subgroups (33). Because model organisms have reduced genetic heterogeneity, we observed that in normal chow–fed animals at 5.5 weeks of age (Fig. 3G), male and female mice solely expressing WT proinsulin (black symbols), Ins2-proinsulin-R(B22)E heterozygotes (blue symbols), and homozygotes (red symbols) tended to fall into three distinct groups when plotting circulating proinsulin levels versus simultaneous random blood glucose. Ins2-proinsulin-R(B22)E heterozygotes showed the greatest variability of circulating proinsulin among mice, which was not well-correlated with random glucose, and overall, these values were nearly normal (Fig. 3G). However, homozygotes formed a discrete group, suggesting a positively sloped relationship of circulating proinsulin with simultaneous random blood glucose. Given the developing insulin deficiency (Fig. 3C), these differences were amplified when considering the circulating proinsulin-to-insulin ratio in homozygotes (Fig. 3H), although only small differences remained between WT and heterozygous animals. (A separate set of heterozygous HFD-fed males at 11 weeks of age [distinct from the animals shown in Fig. 3G and H] progressed into diabetes, yet the mean value for circulating proinsulin or proinsulin-to-insulin ratio was not increased [Supplementary Fig. 2].) Altogether, the data support that circulating proinsulin-to-insulin ratio relative to prevailing glucose does serve to distinguish animal subgroups.

We next examined islet insulin and proinsulin content and localization in fixed tissues. In WT mice, β-cells exhibit robust insulin immunofluorescence, and proinsulin is concentrated in a juxtanuclear subregion (Fig. 4A). In Ins2-proinsulin-R(B22)E heterozygous males, again most islet cells are β-cells exhibiting obvious insulin immunofluorescence, but there were a few cells with diminished insulin signal and more expansive proinsulin immunofluorescence (suggesting depletion of insulin stores yet maintenance of biosynthetic activity [Fig. 4B]). Young Ins2-proinsulin-R(B22)E homozygotes that were not yet diabetic did not initially appear very different from heterozygotes. Animals with random blood glucose <200 mg/dL exhibited a subset of bright insulin-positive cells plus other islet cells with diminished insulin accompanied by increased proinsulin immunofluorescence (Fig. 4C), but this was not a major change (quantified in Fig. 4F). Even after homozygotes developed frank diabetes, there were still a few islet cells brightly immunofluorescent for insulin, but an enlarged fraction of cells showed diminished insulin signal (quantified in Fig. 4F) while exhibiting robust proinsulin immunofluorescence, as seen in both larger and smaller islets (Fig. 4D and E).

Figure 4

Proinsulin and insulin double immunofluorescence showing random blood glucose (BG) at the time of euthanasia. A: WT control. B: Ins2-proinsulin-R(B22)E heterozygote (Het) (6-week-old female). C: Normoglycemic 4-week-old female Ins2-proinsulin-R(B22)E homozygote (Hom). D and E: Ins2-proinsulin-R(B22)E-Hom 7-week-old female. F: Quantitation of insulin-positive cytoplasmic area as a percentage of total β-cell cytoplasmic area in independent islets from the genotypes indicated (mean ± SD; P = 0.0002). ***P < 0.001.

Figure 4

Proinsulin and insulin double immunofluorescence showing random blood glucose (BG) at the time of euthanasia. A: WT control. B: Ins2-proinsulin-R(B22)E heterozygote (Het) (6-week-old female). C: Normoglycemic 4-week-old female Ins2-proinsulin-R(B22)E homozygote (Hom). D and E: Ins2-proinsulin-R(B22)E-Hom 7-week-old female. F: Quantitation of insulin-positive cytoplasmic area as a percentage of total β-cell cytoplasmic area in independent islets from the genotypes indicated (mean ± SD; P = 0.0002). ***P < 0.001.

Close modal

By electron microscopy, β-cells from WT and normoglycemic heterozygous Ins2-proinsulin-R(B22)E mice had a similar, well-granulated appearance (Supplementary Fig. 3A and B). Even in HFD-fed animals, the secretory pathway of WT β-cells exhibited the normal cisternal ER, ER-Golgi vesiculotubular clusters (pre-Golgi intermediates), well-developed Golgi stacks, and immature secretory granules and mature insulin granules (Supplementary Fig. 3C). These features were also present in Ins2-proinsulin-R(B22)E heterozygotes on a normal chow diet (Supplementary Fig. 3A and B). However, on HFD, a β-cell subpopulation in Ins2-proinsulin-R(B22)E heterozygotes developed an expanded ER and underfilled, low-electron-density granule contents (Supplementary Fig. 3D, “cell B”). In homozygotes, it was easy to identify a subset of β-cells bearing some insulin granules, while other β-cells exhibited few granules but increased ER (Fig. 5 and Supplementary Fig. 3E) as well as cells bearing underfilled low-electron-density “microgranules” and related secretory pathway organelles (Supplementary Fig. 3F and G). The changes in granule number and appearance are quantified in Fig. 5C.

Figure 5

Transmission electron microscopy of islet cells from a 4-week-old male Ins2-proinsulin-R(B22)E homozygote (Hom), highlighting heterogeneity in the abundance of insulin secretory granules. A: Lower power view, with β-cells identified. The cell at the bottom and the two cells in the upper left corner exhibit glucagon secretory granules; random blood glucose (BG) was taken at the time of euthanasia. B: The yellow boxed area from A shown at higher power. Vesiculotubular clusters (VTCs) are a pre-Golgi compartment. A more detailed electron microscopy survey is presented in Supplementary Fig. 3. C: For each indicated genotype and diet, total β-cell granule numbers were counted per unit area (25-µm2 boxed areas). Those with a classic electron-dense granule core were considered to be mature (green), and those lacking the classic electron-dense core were considered to be underfilled (red). Quantification compared HFD-fed (H) WT and heterozygous (Het) conditions and normal chow (N) diet–fed Het and Hom conditions. Data are mean ± SD. **P < 0.01, ****P < 0.0001. Mito, mitochondrion.

Figure 5

Transmission electron microscopy of islet cells from a 4-week-old male Ins2-proinsulin-R(B22)E homozygote (Hom), highlighting heterogeneity in the abundance of insulin secretory granules. A: Lower power view, with β-cells identified. The cell at the bottom and the two cells in the upper left corner exhibit glucagon secretory granules; random blood glucose (BG) was taken at the time of euthanasia. B: The yellow boxed area from A shown at higher power. Vesiculotubular clusters (VTCs) are a pre-Golgi compartment. A more detailed electron microscopy survey is presented in Supplementary Fig. 3. C: For each indicated genotype and diet, total β-cell granule numbers were counted per unit area (25-µm2 boxed areas). Those with a classic electron-dense granule core were considered to be mature (green), and those lacking the classic electron-dense core were considered to be underfilled (red). Quantification compared HFD-fed (H) WT and heterozygous (Het) conditions and normal chow (N) diet–fed Het and Hom conditions. Data are mean ± SD. **P < 0.01, ****P < 0.0001. Mito, mitochondrion.

Close modal

Insulin/proinsulin double-immunofluorescence images of heterozygous and homozygous Ins2-proinsulin-R(B22)E animals also revealed some islet cells unlabeled for either marker (Fig. 4B–E). Three-color immunofluorescence in Ins2-proinsulin-R(B22)E-positive islets labeled glucagon-positive cells initially at the perimeter of normoglycemic WT islets (Fig. 6A), which appeared increasingly within the islet interior, and corresponded to most of the remaining cells (Fig. 6B–E), especially in homozygous mice (quantified in Supplementary Fig. 4). Together, the data indicate that while progressing from WT to heterozygous to homozygous diabetic animals, a decrease of insulin-storing β-cells (Figs. 4F and 5C) is seen with increasing proinsulin-enriched cells (Figs. 4 and 6) and glucagon-enriched cells (Supplementary Fig. 4). The loss of insulin in homozygotes was also observed by immunoblotting of islet lysates analyzed by reducing SDS-PAGE. Insulin deficiency was less noticeable when the homozygotes were still at the euglycemic stage but was exacerbated in parallel with the progression of hyperglycemia (Fig. 7A and B).

Figure 6

Growing subpopulations of proinsulin-enriched cells (red) and glucagon-enriched cells (blue) and a decline of insulin-enriched cells (green) in Ins2-proinsulin-R(B22)E heterozygotes (Het) and homozygotes (Hom), as identified by triple immunofluorescence. Random blood glucose levels are indicated. A: WT 6-week-old female. B and C: Ins2-proinsulin-R(B22)E-Het females (age 6.5 weeks). D and E: Ins2-proinsulin-R(B22)E-Hom females (ages 4 and 6 weeks, respectively). Proins, proinsulin.

Figure 6

Growing subpopulations of proinsulin-enriched cells (red) and glucagon-enriched cells (blue) and a decline of insulin-enriched cells (green) in Ins2-proinsulin-R(B22)E heterozygotes (Het) and homozygotes (Hom), as identified by triple immunofluorescence. Random blood glucose levels are indicated. A: WT 6-week-old female. B and C: Ins2-proinsulin-R(B22)E-Het females (age 6.5 weeks). D and E: Ins2-proinsulin-R(B22)E-Hom females (ages 4 and 6 weeks, respectively). Proins, proinsulin.

Close modal
Figure 7

Progressive loss of insulin, accompanied by proinsulin misfolding, in Ins2-proinsulin-R(B22)E homozygotes (Hom). A: Reducing SDS-PAGE and immunoblotting with anti-proinsulin (top panel), anti-insulin (middle panel), and anti-cyclophilin B (CypB) (loading control, bottom panel). Islets were isolated from males (ages 5–7 weeks) with the genotypes shown above; random blood glucose level at the time of euthanasia is also indicated. B: Nonreducing SDS-PAGE and immunoblotting of the identical samples as that shown in A, highlighting aberrant proinsulin disulfide-linked complexes. C: Immunoblotting of p58ipk and BiP from male WT and Ins2-proinsulin-R(B22)E heterozygotes (Het) and Hom (age 5–6 weeks). D: Quantitation of immunoblotting like that shown in C (n = 6 per group). Statistical analysis (mean ± SD) shows that changes in normalized BiP protein levels were nonsignificant. **P < 0.01. Proins, proinsulin.

Figure 7

Progressive loss of insulin, accompanied by proinsulin misfolding, in Ins2-proinsulin-R(B22)E homozygotes (Hom). A: Reducing SDS-PAGE and immunoblotting with anti-proinsulin (top panel), anti-insulin (middle panel), and anti-cyclophilin B (CypB) (loading control, bottom panel). Islets were isolated from males (ages 5–7 weeks) with the genotypes shown above; random blood glucose level at the time of euthanasia is also indicated. B: Nonreducing SDS-PAGE and immunoblotting of the identical samples as that shown in A, highlighting aberrant proinsulin disulfide-linked complexes. C: Immunoblotting of p58ipk and BiP from male WT and Ins2-proinsulin-R(B22)E heterozygotes (Het) and Hom (age 5–6 weeks). D: Quantitation of immunoblotting like that shown in C (n = 6 per group). Statistical analysis (mean ± SD) shows that changes in normalized BiP protein levels were nonsignificant. **P < 0.01. Proins, proinsulin.

Close modal

Remarkably, islets of Ins2-proinsulin-R(B22)E homozygotes did not exhibit proinsulin deficiency, even as hyperglycemia progressed into the 400–500 mg/dL range (Fig. 7A). However, in homozygous Ins2-proinsulin-R(B22)E mice, a greater fraction of proinsulin was contained in aberrant disulfide-linked complexes (Fig. 7B), which have been reported both in stressed human islets and murine diabetes models (17). Indeed, even before development of frank diabetes, we noted a tendency toward increased islet BiP protein (as well as the BiP cochaperone p58ipk [Fig. 7C and D]), although the magnitude of the effect on these downstream targets of ER stress response was only approximately twofold. ER resident proteins (reactive with anti-KDEL) (Supplementary Fig. 5A) became more apparent in proinsulin-enriched cells of heterozygotes (mean ∼30% of β-cells) and homozygotes (mean ∼50% of β-cells) (Supplementary Fig. 5B), consistent with cellular heterogeneity that includes an increased subfraction of β-cells maintaining an expanded or activated ER compartment.

We conducted three independent pulse-chase radiolabeling experiments to look at the efficiency of insulin biosynthesis. First, the amount of newly synthesized proinsulin (made in a 30-min pulse labeling with 35S-amino acids) was analyzed by immunoprecipitation with anti-insulin, reducing SDS-PAGE, and autoradiography (Fig. 8A, right). At 2 h of chase, cells and media were combined together before immunoprecipitation, and the yield of newly made insulin (Fig. 8A, left) derived from labeled proinsulin in WT control islets was nearly 100%, whereas insulin generation in euglycemic homozygous Ins2-proinsulin-R(B22)E islets was less efficient (quantitation in Fig. 8D). In the next experiment, labeled heterozygous Ins2-proinsulin-R(B22)E islets were compared against those from a homozygote with random blood glucose of 400 mg/dL (Fig. 8B). From the euglycemic heterozygote, recovery of labeled insulin at 2-h chase from the original newly synthesized proinsulin was excellent (Fig. 8B, left), approaching 100% (Fig. 8D). However, in the homozygote, little labeled insulin was produced (Fig. 8B), with a yield of only 24% (Fig. 8D). In a third experiment, recovery of mature insulin from newly synthesized proinsulin in a euglycemic heterozygote again approached 100% (Fig. 8C), whereas in a homozygote with a random blood glucose of 537 mg/dL, insulin yield was only 4% (Fig. 8D). Remarkably, in each case, newly synthesized proinsulin in the homozygous Ins2-proinsulin-R(B22)E islets was detected by nonreducing SDS-PAGE as a ladder of aberrant disulfide-linked proinsulin complexes that exceeded the recovery of monomeric proinsulin (Fig. 8A–C). Indeed, recovery of newly synthesized monomeric proinsulin by nonreducing SDS-PAGE worsened with the progression of hyperglycemia, tightly correlating increased proinsulin disulfide-linked complex formation with decreased insulin production (Fig. 8A–C).

Figure 8

Biosynthesis of proinsulin and insulin in WT control and Ins2-proinsulin-R(B22)E heterozygous (Het) and homozygous (Hom) males with progression of diabetes (age 4–8 weeks). The random blood glucose of each animal at the time of euthanasia is indicated. Isolated islets were pulse labeled with 35S-amino acids for 30 min and then chased for 2 h, as indicated. Cells (C) and chase media (M) were either combined (so that no protein was lost) or analyzed separately. Samples (normalized to trichloroacetic acid–precipitable counts in the cell lysates) were immunoprecipitated with anti-insulin followed by Tris-tricine-urea-SDS-PAGE under nonreducing or reducing conditions followed by fluorography. A line is drawn separating the nonreduced and reduced samples, but these images show the complete gels, and no lanes have been excised. AC: Three independent experiments with the genotypes shown (ages 4, 5, and 8 weeks, respectively). D: Quantitative recovery of newly synthesized insulin derived from pulse-labeled proinsulin, as derived from the preceding phosphorimages; genotype and random blood glucose are indicated. Proinsulin bands (at chase time 0) were quantitated from reducing gels; newly synthesized insulin derived from the pulse-labeled samples were quantified from nonreducing gels. (Insulin is a two-chain protein that “falls apart” under reducing conditions; thus, nonreducing gels are preferable for this analysis.) ox, oxidized; R, reduced.

Figure 8

Biosynthesis of proinsulin and insulin in WT control and Ins2-proinsulin-R(B22)E heterozygous (Het) and homozygous (Hom) males with progression of diabetes (age 4–8 weeks). The random blood glucose of each animal at the time of euthanasia is indicated. Isolated islets were pulse labeled with 35S-amino acids for 30 min and then chased for 2 h, as indicated. Cells (C) and chase media (M) were either combined (so that no protein was lost) or analyzed separately. Samples (normalized to trichloroacetic acid–precipitable counts in the cell lysates) were immunoprecipitated with anti-insulin followed by Tris-tricine-urea-SDS-PAGE under nonreducing or reducing conditions followed by fluorography. A line is drawn separating the nonreduced and reduced samples, but these images show the complete gels, and no lanes have been excised. AC: Three independent experiments with the genotypes shown (ages 4, 5, and 8 weeks, respectively). D: Quantitative recovery of newly synthesized insulin derived from pulse-labeled proinsulin, as derived from the preceding phosphorimages; genotype and random blood glucose are indicated. Proinsulin bands (at chase time 0) were quantitated from reducing gels; newly synthesized insulin derived from the pulse-labeled samples were quantified from nonreducing gels. (Insulin is a two-chain protein that “falls apart” under reducing conditions; thus, nonreducing gels are preferable for this analysis.) ox, oxidized; R, reduced.

Close modal

We report that unlike Akita-proinsulin-C(A7)Y, proinsulin-R(B22)E can pass ER quality control to become secreted from Min6 pancreatic β-cells (Fig. 1B), and its expression does not efficiently block export of coexpressed WT proinsulin in these cells (Fig. 1C). Moreover, in a normal laboratory environment, heterozygous Ins2-proinsulin-R(B22)E males gained weight postweaning and exhibited normal glucose tolerance, and diabetes did not develop (Fig. 2A–D) for as long as we followed the animals (up to 6 months of life). Nevertheless, different cell lines may exhibit different degrees of ER quality control of mutant proinsulin (34); moreover, proinsulin-R(B22)E showed evidence of misfolding in the ER (Figs. 1, 7, and 8, and Supplementary Fig. 1A). Additionally, when transitioned to an HFD, all male heterozygotes developed progressive hyperglycemia/diabetes (Fig. 2D–F) with an inadequate insulin secretory response (Fig. 2G and H) because of diminished islet insulin content (Fig. 2J), accounting for impaired GSIS (Fig. 2I) despite the persistence of proinsulin (Fig. 2J and Supplementary Fig. 1B) in a growing percentage of β-cells (Fig. 4B). The pathological effects of HFD may include contributions from 1) increased insulin demand that exceeds insulin storage, 2) increased proinsulin misfolding secondary to upregulated proinsulin biosynthesis, and/or 3) adverse effects on ER homeostatic function secondary to nutrients contained within HFD.

The sexually dimorphic effects of proinsulin misfolding seen in the heterozygotes is amply documented in mouse models of MIDY as well as several other forms of diabetes. Akita mice, which bear one allele encoding misfolded proinsulin-C(A7)Y, exhibit a selective diabetes phenotype in males (35). A similar sexual dimorphism is observed in Kuma mice bearing proinsulin-Q(A15)del (36). Male KINGS mice bearing one allele encoding proinsulin-G(B8)S are overtly diabetic at ∼5 weeks, whereas females have only slightly elevated nonfasting glycemia (6). Furthermore, in Munich mice bearing proinsulin-C(A6)S, even at 6 months of age, males have diabetes (by fasting glucose criteria) and females do not (37). One explanation may be that estrogen helps to promote the successful degradation of mutant proinsulin (38). Interestingly, even in the absence of any Ins gene mutations, low-dose streptozotocin injections in five different strains of mice (C57BL/6, MRL/Mp, BALB/c, DBA/2, and 129/SvEv) results in males developing diabetes to a greater extent than females (35). Our current studies of Ins2-proinsulin-R(B22)E mice are thus consistent with the established sexually dimorphic hyperglycemia phenotype in many murine models.

A similar phenotype develops in male and female homozygotes even without HFD (Figs. 3A–F and 4C–E). An increase of circulating proinsulin-to-insulin ratio has been described in human T2D (39,40); an intra-islet increase of proinsulin-to-insulin has also been reported during development of spontaneous T2D in animal models (41). We also observed an increasing proinsulin-to-insulin ratio in the circulation (Fig. 3H) and in the islets themselves (Figs. 4C–E and 7A) in diabetic homozygotes.

Proinsulin misfolding is causal for diabetes in the homozygotes, with obviously aberrant disulfide-linked proinsulin complexes forming immediately upon synthesis and accompanying diminished insulin biosynthesis (Figs. 7B and 8). Whereas the ability of isolated islets from WT versus heterozygous versus homozygous mice to respond to a glucose challenge exhibits no obvious defect in fold stimulation, a progressive deficiency of insulin secretion under both unstimulated and stimulated conditions is apparent in vivo and in vitro (Figs. 2I and 3E and F), consistent with a failure to maintain the insulin storage pool (42).

The progression of dysglycemia/diabetes is linked to the emergence of intra-islet heterogeneity, with increasing proinsulin-rich cells (4346) bearing little or no stored insulin—distinct from the subpopulation of β-cells rich in stored insulin (Fig. 4). Heterogeneity within the β-cell population is increasingly recognized at the mRNA level (47), including the description of “extreme” β-cells with lower insulin levels and higher juxtanuclear proinsulin immunostaining (48), which is believed to represent the Golgi region from which new secretory granules emerge (23,49). However, the diabetogenic progression in HFD-challenged heterozygous Ins2-proinsulin-R(B22)E mice (or homozygotes) involves islet cells exhibiting a proinsulin localization that fills the cytoplasm (Fig. 4) with expansion of the ER (Fig. 5 and Supplementary Fig. 3) and with increased generation of misfolded proinsulin (Figs. 7 and 8) accompanied by increased KDEL-containing ER resident proteins (Supplementary Fig. 5) and deficient insulin biosynthesis (Fig. 8) with deficient insulin content (Fig. 2J). Curiously, this is what has been observed for the localization and misfolding of proinsulin in islets of T2D-like mice with hyperphagia-induced dysglycemia without any Ins gene variant (17). The total life span of proinsulin in β-cells is limited to ∼4 h (23), suggesting that β-cells lacking insulin but bearing proinsulin (which may be entirely overlooked in β-cell mass measurements [50]) remain biosynthetically active right up to the time of our analysis.

Additionally, even in “off-scale high-glucose” Ins2-proinsulin-R(B22)E homozygotes, a subset of β-cells with substantial stored insulin content and only modest proinsulin persists, although they represent a shrinking fraction of the total (Figs. 4 and 6). Conceivably, such cells could represent immature β-cells with deficient glucose sensing (51,52) that may not release stored insulin, but this remains to be determined. Additionally, the diseased islets develop increased glucagon-positive cells within the islet interior (Fig. 6), a feature noted in several diabetes models. Altogether, these data are consistent with the existence of islet β-cell subpopulations exhibiting heterogeneity in ER homeostasis (53) and an increase in islet α-to-β-cell ratio (54).

In summary, the main observation in this article is that proinsulin misfolding can be entirely subclinical, yet dramatic pathology emerges upon HFD exposure, triggering rapid insulin deficiency. HFD-induced β-cell failure has been proposed to be alternatively associated with gluco/lipotoxicity, β-cell senescence, dedifferentiation, transdifferentiation, or apoptosis (55). This article does not resolve those alternatives but highlights intra-islet heterogeneity, with decreasing insulin-high/proinsulin-low cells, increasing proinsulin-high/insulin-low cells, and glucagon-positive cells during disease progression. Unequivocally, our data in these models show that development of hyperglycemia runs antiparallel with pancreatic insulin storage (i.e., biosynthesis of new insulin secretory granules is inadequate to replace the depletion of stored insulin used to meet the body’s metabolic needs. Fascinating work is ongoing worldwide to understand the changes in islet cell heterogeneity during the development of diabetes (5355). We merely emphasize that all the pathological changes identified herein can be triggered by a genetic predisposition to proinsulin misfolding. Therefore, we conclude that predisposition to proinsulin misfolding serves as an important potential risk factor to diet-induced diabetes.

This article contains supplementary material online at https://doi.org/10.2337/figshare.15173610.

Acknowledgments. We thank Leroux Devon, Michigan Biomedical Research Electron Microscopy Core Facility, for support and the Michigan Tissue and Molecular Pathology (Histology) Core for help with sample preparation.

Funding. This work was supported by National Institutes of Health grants R01-DK-48280 and P01-AI-118688, the Michigan Diabetes Research Center Morphology Core (P30-DK-020572), the University of Michigan and Protein Folding Diseases Initiative, and Howard Hughes Medical Institute, University of Colorado Anschutz Medical School, and National Jewish Health institutional funds.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. M.A., A.A., L.H., M.T., and N.J. generated research data, reviewed data, and contributed to discussion. D.L. developed analysis techniques. J.K. and P.A. initiated and designed the research project. N.J. collaborated with Dr. Jennifer Matsuda and the team in the National Jewish Mouse Genetics Core Facility in the design and creation of the original NOD knockin mouse that gave rise to the mice used in these studies. P.A. wrote the manuscript. All authors edited and reviewed the manuscript. P.A. 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 a symposium talk at the 81st Scientific Sessions of the American Diabetes Association, 25–29 June 2021.

1
Liu
M
,
Hodish
I
,
Haataja
L
, et al
.
Proinsulin misfolding and diabetes: mutant INS gene-induced diabetes of youth
.
Trends Endocrinol Metab
2010
;
21
:
652
659
2
Sun
J
,
Cui
J
,
He
Q
,
Chen
Z
,
Arvan
P
,
Liu
M
.
Proinsulin misfolding and endoplasmic reticulum stress during the development and progression of diabetes
.
Mol Aspects Med
2015
;
42
:
105
118
3
Arunagiri
A
,
Haataja
L
,
Cunningham
CN
, et al
.
Misfolded proinsulin in the endoplasmic reticulum during development of beta cell failure in diabetes
.
Ann N Y Acad Sci
2018
;
1418
:
5
19
4
Arneth
B
.
Insulin gene mutations and posttranslational and translocation defects: associations with diabetes
.
Endocrine
2020
;
70
:
488
497
5
Blutke
A
,
Renner
S
,
Flenkenthaler
F
, et al
.
The Munich MIDY Pig Biobank–a unique resource for studying organ crosstalk in diabetes
.
Mol Metab
2017
;
6
:
931
940
6
Austin
ALF
,
Daniels Gatward
LF
,
Cnop
M
, et al
.
The KINGS Ins2+/G32S mouse: a novel model of β-cell endoplasmic reticulum stress and human diabetes
.
Diabetes
2020
;
69
:
2667
2677
7
Sprunger
ML
,
Jackrel
ME
.
Quality control in the ER: misfolded prohormones get a checkup
.
Mol Cell
2019
;
75
:
415
416
8
Kim
YH
,
Kastner
K
,
Abdul-Wahid
B
,
Izaguirre
JA
.
Evaluation of conformational changes in diabetes-associated mutation in insulin a chain: a molecular dynamics study
.
Proteins
2015
;
83
:
662
669
9
Ionescu-Tirgoviste
C
,
Despa
F
.
Biophysical alteration of the secretory track in β-cells due to molecular overcrowding: the relevance for diabetes
.
Integr Biol
2011
;
3
:
173
179
10
Hodish
I
,
Liu
M
,
Rajpal
G
, et al
.
Misfolded proinsulin affects bystander proinsulin in neonatal diabetes
.
J Biol Chem
2010
;
285
:
685
694
11
Sun
J
,
Xiong
Y
,
Li
X
, et al
.
Role of proinsulin self-association in mutant INS gene-induced diabetes of youth
.
Diabetes
2020
;
69
:
954
964
12
Barbetti
F
,
Mammì
C
,
Liu
M
, et al
.
Neonatal diabetes: permanent neonatal diabetes and transient neonatal diabetes
. In
Diabetes associated with single gene defects and chromosomal abnormalities
.
Barbetti
F
,
Ghizzoni
L
,
Guaraldi
F
, Eds.
Basel, Switzerland
,
Karger AG
,
2017
, pp.
1
25
13
Liu
M
,
Sun
J
,
Cui
J
, et al
.
INS-gene mutations: from genetics and beta cell biology to clinical disease
.
Mol Aspects Med
2015
;
42
:
3
18
14
Steiner
DF
.
The proinsulin C-peptide–a multirole model
.
Exp Diabesity Res
2004
;
5
:
7
14
15
Irwin
DM
.
Evolution of the mammalian insulin (Ins) gene; changes in proteolytic processing
.
Peptides
2021
;
135
:
170435
16
Rege
NK
,
Liu
M
,
Yang
Y
, et al
.
Evolution of insulin at the edge of foldability and its medical implications
.
Proc Natl Acad Sci U S A
2020
;
117
:
29618
29628
17
Arunagiri
A
,
Haataja
L
,
Pottekat
A
, et al
.
Proinsulin misfolding is an early event in the progression to type 2 diabetes
.
eLife
2019
;
8
:
e44532
18
Støy
J
,
Olsen
J
,
Park
SY
,
Gregersen
S
,
Hjørringgaard
CU
,
Bell
GI
.
In vivo measurement and biological characterisation of the diabetes-associated mutant insulin p.R46Q (GlnB22-insulin)
.
Diabetologia
2017
;
60
:
1423
1431
19
Molven
A
,
Ringdal
M
,
Nordbø
AM
, et al.;
Norwegian Childhood Diabetes Study Group
.
Mutations in the insulin gene can cause MODY and autoantibody-negative type 1 diabetes
.
Diabetes
2008
;
57
:
1131
1135
20
Rose
K
,
Rees
AR
,
Drake
CS
,
Offord
RE
.
The role of the arginine-B22 residue in insulin action
.
Biochem J
1981
;
195
:
765
768
21
Zhu
SQ
,
Li
TF
,
Cui
DF
,
Cao
QP
,
Zhang
YS
.
Effect of B22-arginine replacement on the biological activity of insulin
.
Sci Sin
1981
;
24
:
264
271
22
Du
H-J
,
Shi
J-H
,
Cui
D-F
,
Zhang
Y-S
.
B22 Glu des-B30 insulin: a novel monomeric insulin
.
Acta Biochim Biophys Sin (Shanghai)
2006
;
38
:
537
542
23
Haataja
L
,
Snapp
E
,
Wright
J
, et al
.
Proinsulin intermolecular interactions during secretory trafficking in pancreatic β cells
.
J Biol Chem
2013
;
288
:
1896
1906
24
Liu
M
,
Lara-Lemus
R
,
Shan
SO
, et al
.
Impaired cleavage of preproinsulin signal peptide linked to autosomal-dominant diabetes
.
Diabetes
2012
;
61
:
828
837
25
Miyazaki
J
,
Araki
K
,
Yamato
E
, et al
.
Establishment of a pancreatic beta cell line that retains glucose-inducible insulin secretion: special reference to expression of glucose transporter isoforms
.
Endocrinology
1990
;
127
:
126
132
26
Hua
QX
,
Liu
M
,
Hu
SQ
,
Jia
W
,
Arvan
P
,
Weiss
MA
.
A conserved histidine in insulin is required for the foldability of human proinsulin: structure and function of an ALAB5 analog
.
J Biol Chem
2006
;
281
:
24889
24899
27
Liu
M
,
Wan
ZL
,
Chu
YC
, et al
.
Crystal structure of a “nonfoldable” insulin: impaired folding efficiency despite native activity
.
J Biol Chem
2009
;
284
:
35259
35272
28
Liu
M
,
Hua
QX
,
Hu
SQ
, et al
.
Deciphering the hidden informational content of protein sequences: foldability of proinsulin hinges on a flexible arm that is dispensable in the mature hormone
.
J Biol Chem
2010
;
285
:
30989
31001
29
Rege
NK
,
Liu
M
,
Dhayalan
B
, et al
.
“Register-shift” insulin analogs uncover constraints of proteotoxicity in protein evolution
.
J Biol Chem
2020
;
295
:
3080
3098
30
Porte
D
Jr.
,
Kahn
SE
.
β-Cell dysfunction and failure in type 2 diabetes: potential mechanisms
.
Diabetes
2001
;
50
(
Suppl. 1
):
S160
S163
31
Haffner
SM
,
Gonzalez
C
,
Mykkänen
L
,
Stern
M
.
Total immunoreactive proinsulin, immunoreactive insulin and specific insulin in relation to conversion to NIDDM: the Mexico City Diabetes Study
.
Diabetologia
1997
;
40
:
830
837
32
Egan
AM
,
Laurenti
MC
,
Hurtado Andrade
MD
, et al
.
Limitations of the fasting proinsulin to insulin ratio as a measure of β-cell health in people with and without impaired glucose tolerance
.
Eur J Clin Invest
2021
;
51
:
e13469
33
Li
T
,
Quan
H
,
Zhang
H
, et al
.
Subgroup analysis of proinsulin and insulin levels reveals novel correlations to metabolic indicators of type 2 diabetes
.
Aging (Albany N Y)
2020
;
12
:
10715
10735
34
Haataja
L
,
Arunagiri
A
,
Hassan
A
, et al
.
Distinct states of proinsulin misfolding in MIDY
.
Cell Mol Life Sci
2021
;
78
:
6017
6031
35
Gurley
SB
,
Clare
SE
,
Snow
KP
,
Hu
A
,
Meyer
TW
,
Coffman
TM
.
Impact of genetic background on nephropathy in diabetic mice
.
Am J Physiol Renal Physiol
2006
;
290
:
F214
F222
36
Sakano
D
,
Inoue
A
,
Enomoto
T
, et al
.
Insulin2Q104del (Kuma) mutant mice develop diabetes with dominant inheritance
.
Sci Rep
2020
;
10
:
12187
37
Herbach
N
,
Rathkolb
B
,
Kemter
E
, et al
.
Dominant-negative effects of a novel mutated Ins2 allele causes early-onset diabetes and severe beta-cell loss in Munich Ins2C95S mutant mice
.
Diabetes
2007
;
56
:
1268
1276
38
Xu
B
,
Allard
C
,
Alvarez-Mercado
AI
, et al
.
Estrogens promote misfolded proinsulin degradation to protect insulin production and delay diabetes
.
Cell Rep
2018
;
24
:
181
196
39
Røder
ME
,
Porte
D
Jr.
,
Schwartz
RS
,
Kahn
SE
.
Disproportionately elevated proinsulin levels reflect the degree of impaired B cell secretory capacity in patients with noninsulin-dependent diabetes mellitus
.
J Clin Endocrinol Metab
1998
;
83
:
604
608
40
Prentki
M
,
Nolan
CJ
.
Islet beta cell failure in type 2 diabetes
.
J Clin Invest
2006
;
116
:
1802
1812
41
Yang
K
,
Gotzmann
J
,
Kuny
S
,
Huang
H
,
Sauvé
Y
,
Chan
CB
.
Five stages of progressive β-cell dysfunction in the laboratory Nile rat model of type 2 diabetes
.
J Endocrinol
2016
;
229
:
343
356
42
Liu
M
,
Huang
Y
,
Xu
X
, et al
.
Normal and defective pathways in biogenesis and maintenance of the insulin storage pool
.
J Clin Invest
2021
;
131
:
e142240
43
Schuit
FC
,
In’t Veld
PA
,
Pipeleers
DG
.
Glucose stimulates proinsulin biosynthesis by a dose-dependent recruitment of pancreatic beta cells
.
Proc Natl Acad Sci U S A
1988
;
85
:
3865
3869
44
Kiekens
R
,
In’t Veld
P
,
Mahler
T
,
Schuit
F
,
Van De Winkel
M
,
Pipeleers
D
.
Differences in glucose recognition by individual rat pancreatic B cells are associated with intercellular differences in glucose-induced biosynthetic activity
.
J Clin Invest
1992
;
89
:
117
125
45
Pipeleers
D
,
Kiekens
R
,
Ling
Z
,
Wilikens
A
,
Schuit
F
.
Physiologic relevance of heterogeneity in the pancreatic beta-cell population
.
Diabetologia
1994
;
37
(
Suppl. 2
):
S57
S64
46
Bosco
D
,
Meda
P
.
Actively synthesizing beta-cells secrete preferentially after glucose stimulation
.
Endocrinology
1991
;
129
:
3157
3166
47
Xin
Y
,
Dominguez Gutierrez
G
,
Okamoto
H
, et al
.
Pseudotime ordering of single human β-cells reveals states of insulin production and unfolded protein response
.
Diabetes
2018
;
67
:
1783
1794
48
Farack
L
,
Golan
M
,
Egozi
A
, et al
.
Transcriptional heterogeneity of beta cells in the intact pancreas
.
Dev Cell
2019
;
48
:
115
125.e4
49
Orci
L
,
Ravazzola
M
,
Amherdt
M
,
Madsen
O
,
Vassalli
JD
,
Perrelet
A
.
Direct identification of prohormone conversion site in insulin-secreting cells
.
Cell
1985
;
42
:
671
681
50
Oram
RA
,
Sims
EK
,
Evans-Molina
C
.
Beta cells in type 1 diabetes: mass and function; sleeping or dead?
Diabetologia
2019
;
62
:
567
577
51
Beamish
CA
,
Zhang
L
,
Szlapinski
SK
,
Strutt
BJ
,
Hill
DJ
.
An increase in immature β-cells lacking Glut2 precedes the expansion of β-cell mass in the pregnant mouse
.
PLoS One
2017
;
12
:
e0182256
52
van der Meulen
T
,
Mawla
AM
,
DiGruccio
MR
, et al
.
Virgin beta cells persist throughout life at a neogenic niche within pancreatic islets
.
Cell Metab
2017
;
25
:
911
926.e6
53
Wang
YJ
,
Kaestner
KH
.
Single-cell RNA-seq of the pancreatic islets–a promise not yet fulfilled?
Cell Metab
2019
;
29
:
539
544
54
Gromada
J
,
Chabosseau
P
,
Rutter
GA
.
The α-cell in diabetes mellitus
.
Nat Rev Endocrinol
2018
;
14
:
694
704
55
Hudish
LI
,
Reusch
JE
,
Sussel
L
.
β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes
.
J Clin Invest
2019
;
129
:
4001
4008
56
Soleimanpour
SA
,
Gupta
A
,
Bakay
M
, et al
.
The diabetes susceptibility gene Clec16a regulates mitophagy
.
Cell
2014
;
157
:
1577
1590
57
Liu
M
,
Haataja
L
,
Wright
J
, et al
.
Mutant INS-gene induced diabetes of youth: proinsulin cysteine residues impose dominant-negative inhibition on wild-type proinsulin transport
.
PLoS One
2010
;
5
:
e13333
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/content/license.