Abstract
In pancreatic β cells, misfolded proinsulin is a substrate for ER-associated protein degradation (ERAD) via HRD1/SEL1L. Alternately, β cell HRD1 activity is reported to improve, or impair, insulin biogenesis. Further, while β cell SEL1L deficiency causes HRD1 hypofunction and diminishes islet insulin content, reports conflict as to whether β cell ERAD deficiency increases or decreases proinsulin levels. Here, we examined β cell–specific Hrd1-KO mice (chronic deficiency) and rodent (and human islet) β cells treated acutely with HRD1 inhibitor. β-Hrd1–KO mice developed diabetes with decreased islet proinsulin, yet a relative increase of misfolded proinsulin redistributed to the ER. They also showed upregulated biochemical markers of β cell ER stress and autophagy, electron microscopy evidence of ER enlargement and decreased insulin granule content, and increased glucagon-positive islet cells. Misfolded proinsulin was also increased in islets treated with inhibitors of lysosomal degradation. Preceding any loss of total proinsulin, acute HRD1 inhibition triggered increased nonnative proinsulin, increased phospho-eIF2α with inhibited proinsulin synthesis, and increased LC3b-II (the abundance of which requires expression of ΣR1). We posit a subset of proinsulin molecules undergo HRD1-mediated disposal. When HRD1 is unavailable, misfolded proinsulin accumulates, accompanied by increased phospho-eIF2α that limits further proinsulin synthesis, plus ΣR1-dependent autophagy activation, ultimately lowering steady-state β cell proinsulin (and insulin) levels and triggering diabetes.
Keywords: Cell biology, Endocrinology
Keywords: Beta cells, Insulin
We limited ERAD capacity in β-cell-specific HRD1-KO mice, or HRD1 inhibitor-treated β-cells; predisposing to ER stress, diminished proinsulin synthesis, and SigmaR1-dependent activation of autophagy.
Introduction
Misfolded mutant or WT proinsulin has long been posited to be a substrate of ER-associated degradation (ERAD) (1–4) via the SEL1L/HRD1/p97 pathway (5–7) leading to proteasomal degradation (8, 9). In this pathway, HRD1 participates in retrotranslocation of ERAD substrates from the ER lumen, coupled with substrate ubiquitylation (10); SEL1L participates as a HRD1 partner protein and cofactor (11); and p97 participates in extraction of ubiquitylated substrates from the ER membrane for delivery to proteasomes in the cytosol (12).
In the last 6 years, studies have begun to probe the importance of this pathway for proper pancreatic β cell function. Despite considerable effort, confusion has remained over the directionality of the impact of increased or decreased ERAD function in pancreatic β cells and its implications for the development of diabetes. One group has reported that increased β cell expression of HRD1 (such as may occur in type 2 diabetes or in mouse models of the disease) triggers impaired insulin secretion, whereas impaired ERAD capacity by HRD1 knockdown (KD) improves blood glucose control (13). However, a state of HRD1 KD exists in all cells upon diminished expression of SEL1L — its interaction confers HRD1 stability in mammalian cells (11, 14, 15) and in yeast (16) — but rather than improving glucose-stimulated insulin secretion (GSIS) and blood glucose homeostasis, deficient ERAD capacity was found to result in diminished GSIS with development of whole animal glucose intolerance (6). Indeed, mice genetically deficient for β cell expression of Sel1L have been reported to have normal embryonic development of islets but proceed to severe diabetes with evidence of altered islet cell transcriptional states (17), consistent with islet cell heterogeneity that has been described in several models of full-blown diabetes in rodents (and in humans) (18). Yet, there remains considerable discrepancy even between the 2 groups that have been studying deficient ERAD capacity caused by β cell Sel1L deletion. Both groups identified insulin deficiency, but one found that it was accompanied by a large steady-state increase of β cell proinsulin (6), while the other found a marked decrease in proinsulin content (19). Both studies were performed in the same mouse genetic background, with the only difference being the use of a transgenic RIP-Cre in the first ERAD-deficient model (6) rather than a knockin Ins1-Cre in the other (17, 19).
Here, we sought to clarify the impact of ERAD capacity and report to our knowledge the first β cell–specific deletion of Hrd1 (also known as Synv1), along with appropriate controls, demonstrating that islet proinsulin levels (as well as insulin levels) are unequivocally low in β-Hrd1-KO mice. (We’ve intentionally used the transgenic RIP-Cre; thus, this cannot be the explanation for prior discrepancies.) We find that, beyond the islet cell heterogeneity appearing in the islets of β-Hrd1-KO mice that consistently develop hypoinsulinemic diabetes, there is a notable increase in the phosphorylation state of eIF2α, which is known to limit proinsulin biosynthesis, as well as an activation of autophagy that participates in proinsulin turnover. These effects occurring upon chronic adaptation to β cell–specific loss of HRD1-mediated ERAD capacity are also recapitulated within as little as 2 h of acute chemical HRD1 inhibition, even before any loss of total proinsulin is detected, and the extent of autophagy activation is dependent upon the expression of the ER membrane protein encoded by SigmaR1 (ΣR1), which has recently been identified as a required element for biosynthesis of the critical autophagy factor LC3 (20). Thus, in addition to effects on the differentiation state of pancreatic β cells, ERAD capacity links clearance of misfolded proinsulin to biosynthesis of new proinsulin and the extent of autophagy activation, ultimately regulating insulin content and the risk of developing insulin deficiency and diabetes.
Results
β Cell proinsulin content is diminished in β-Hrd1-KO islets.
We generated mice with β cell–specific deletion of Hrd1 (official gene name: Synv1) using a RIP-Cre transgene (21). To exclude off-target effects (22), we used RIP-Cre–positive Hrd1 heterozygotes as controls. As islets are not purely β cells, the result of immunoblotting β-Hrd1–KO islets is compatible with complete loss of β cell HRD1 expression accompanied by a notable decrease of islet insulin (a representative blot is shown in Supplemental Figure 1A; quantitation of insulin content is shown by ELISA in Supplemental Figure 1B; supplemental material available online with this article; https://doi.org/10.1172/JCI187341DS1) as well as an increase of IRE1α and SEL1L (Supplemental Figure 1C), as reported in other HRD1-deficient cell types (23). β-Hrd1–KO mice did not show a decrease of islet size relative to controls (Supplemental Figure 2) but developed glucose intolerance in males (Figure 1A), females (Figure 1B), and both combined (Figure 1C). Impaired glucose tolerance was coupled with inadequate insulin secretion in vivo (Figure 1D), and when considered as a fraction of (the low) islet insulin content (Supplemental Figure 1B), glucose-augmented insulin secretion (Supplemental Figure 1D) was not decreased from isolated islets of β-Hrd1–KO mice. Nevertheless, within the first 6 postnatal weeks, the body weight of β-Hrd1–KO mice began to grow at a diminished rate compared with controls (Supplemental Figure 3A) and the animals developed random hyperglycemia (Figure 1E). Nevertheless, serum insulin did not rise in response to the increased blood glucose (in fact, it declined), such that the serum insulin/glucose ratio dropped without ever going through a transient hyperinsulinemic period (Supplemental Figure 3B).
Figure 1. Intraperitoneal glucose tolerance test in 6- to 8-week-old β-Hrd1–KO or control mice on normal chow.
(A) Males (mean ± SD). (B) Females (mean ± SD). (C) Combined males + females from A and B. (D) GSIS (control group, 4 males; β-Hrd1–KO, 3 males + 1 female; mean ± SD). (E) Random blood glucose measurements as a function of age in randomly selected males plus females (up to 10 per group). Data were analyzed by 2-way ANOVA with Šidák’s multiple-comparison test; **P < 0.01, ***P < 0.001, ****P < 0.0001.
Although proinsulin is an ERAD substrate, we did not observe the increase in islet proinsulin content previously reported by one group using RIP-Cre to generate ERAD deficiency in β-Sel1L–KO mice (6). Using heterozygous littermate controls, side-by-side comparisons demonstrated proinsulin (and insulin) deficiency (Figure 2A, lane 3, quantitation in Figure 2, C and D), which worsened upon development of diabetes in β-Hrd1–KO mice (Figure 2E), although rarely (using a preweaning β-Hrd1–KO animal) islet proinsulin (and insulin) levels appeared similar to those of control animals (e.g., Figure 2B, lanes 5 and 8). In the islets of β-Hrd1–KO mice with full-blown diabetes, both islet proinsulin and insulin were always low (Figure 2B, lanes 6 and 9). Additionally, islets of β-Hrd1–KO mice stored less IAPP, as detected by quantitative Western blotting (Supplemental Figure 3C), indicating that the phenotype attributable to a deficiency of β cell ERAD capacity is not selective for insulin alone.
Figure 2. Proinsulin and insulin deficiency develops in β-Hrd1–KO mice.
(A) Immunoblotting after SDS-PAGE of islet lysates from control or β-Hrd1–KO male mice (lanes 1–3, random blood glucose values are indicated at top). Reducing gel (above) highlights total proinsulin level (CypB is a loading control); nonreducing gel (below) highlights total mature (2-chain) insulin. (B) Immunoblotting after SDS-PAGE of islet lysates from control or β-Hrd1–KO female mice (lanes 4–6 or 7–9; random blood glucose values indicated at top). Reducing gel (above) highlights total proinsulin level (CypB is a loading control); LC3b is shown below in lanes 7–9. Nonreducing gel highlights nonnative proinsulin molecules in disulfide-linked complexes (above in lanes 4–6); nonreducing gel (below) highlights total mature insulin. (C) Quantitation of immunoblots indicating relative proinsulin content (normalized to HSP90) in islets of β-Hrd1–KO mice (each point represents a different animal; squares = males, circles = females; n is indicated). (D) Quantitation of immunoblots indicating relative content of mature insulin (normalized to HSP90) in islets of β-Hrd1–KO mice (each point represents a different animal; squares = males, circles = females; total n is indicated). Data in C and D were analyzed by unpaired 2-tailed t test; *P < 0.05, **P < 0.01. (E) The data in D and E were replotted relative to the random blood glucose of each animal at the time of euthanasia (proinsulin = closed squares; insulin = open squares).
Impact of β-Hrd1–KO on islet morphology.
In control islets, proinsulin is concentrated in a juxtanuclear pattern (Figure 3A) thought to represent the Golgi region where immature secretory granules are formed (24, 25). However, in β-Hrd1–KO islets, proinsulin tended to have a more diffuse cytoplasmic distribution (Figure 3D), similar to that of the ER (Supplemental Figure 4A). Moreover, ALDH1A3 immunofluorescence (Figure 3B) increased in the islets of β-Hrd1–KO mice (Figure 3E) — an indicator of pancreatic islet dedifferentiation (17, 26, 27) — although some of these cells still retained expression of proinsulin and insulin protein (Supplemental Figure 4B). Additionally in β-Hrd1–KO islets, there was a notable increase in the fraction of glucagon-positive cells (Figure 3, F vs. C). This increase was accompanied by a fractional decrease of insulin-positive cells (Figure 3E, quantified in Figure 3G) and a higher fraction of β cells that exhibited weaker insulin immunostaining, consistent with a decrease of islet insulin (Figure 2D and Supplemental Figure 1B) and an increase in islet cell heterogeneity suggested to be linked to diabetes in humans and several rodent models (18, 28–33).
Figure 3. Proinsulin, insulin, ALDH1A3, and glucagon in pancreatic tissue sections from β-Hrd1–KO mice.
(A and D) Proinsulin immunofluorescence from control and β-Hrd1–KO, respectively (control group, n = 9; β-Hrd1–KO, n = 7). (B and E) ALDH1A3 and insulin from control and β-Hrd1–KO, respectively (control group, n = 4; β-Hrd1–KO, n = 7). (C and F) Glucagon and insulin immunofluorescence from control and β-Hrd1–KO, respectively. Random blood glucose (BG) values are indicated at the bottom of each image. Scale bars: 20 μm. (G) The fraction of glucagon-positive cells versus cells positive for either proinsulin or insulin (sum of both, with each β cell counted only once) from control (random blood glucose mean = 97 mg/dL) and β-Hrd1–KO (random blood glucose mean = 257 mg/dL). For quantitation, the 2 types of islet cells together are referred to as 100% (n = 4 animals per group, each point represents islets in 1 section; unpaired 2-tailed t test, **P < 0.01).
The basis for diminished insulin immunostaining was made clearer upon examination by transmission electron microscopy. At low magnification, insulin granules in the β cells of β-Hrd1–KO islets were hard to discern (Figure 4A). At a higher magnification, it was clear that in both females (Figure 4B) and males (Figure 4C), the diameters of individual insulin secretory granules in β cells from β-Hrd1–KO mice are approximately 55% smaller in size than their control counterparts (Supplemental Figure 5; corresponding to a reduction of cross-sectional area of individual granules of ~80% and a reduction of spherical volume of ~90%). Such microgranules have been reported in other diabetes models, where they are associated with diminished β cell insulin content (34, 35). Moreover, autophagosomes were detected in the cytoplasm (ATG, Figure 4B) and the ER was distended (as highlighted in Figure 4B), suggesting ER stress.
Figure 4. Electron microscopy of β-Hrd1–KO islet sections.
(A) Low-power transmission electron microscopy images of islet tissue sections from control and β-Hrd1–KO females (random blood glucose indicated above). The α and β cells are marked accordingly. (B) Higher magnification of islet sections from mice in A. (C) Higher magnification of islet sections from control and β-Hrd1–KO males (random blood glucose indicated above). Images highlight differences in secretory granule size between control and β-Hrd1–KO animals; ER and autophagosome (ATG) are indicated in B (n = 3 animals per group; quantitation of secretory granule size is shown in Supplemental Figure 5). Scale bars: 1 μm (A), 200 nm (B and C).
ER response in β-Hrd1–KO islets.
Even before the development of diabetes, islets of β-Hrd1–KO mice exhibited significant elevation of the ER luminal chaperone, BiP, and cochaperone, p58ipk (Figure 5A, quantified in Figure 5B). Moreover, β-Hrd1–KO islets exhibited an increase of phospho-eIF2α (Figure 5A, quantified in Figure 5B), which is known to limit proinsulin synthesis (36–39). To check this, we performed metabolic pulse-chase studies with 35S–amino acids and observed a decrease in proinsulin biosynthesis (as well as subsequent generation of mature insulin) in β-Hrd1–KO islets (Supplemental Figure 6A). However, when normalized to TCA-precipitable cpm, the magnitude of the decrease in proinsulin biosynthesis amounted to only 15%, whereas the steady-state proinsulin level decreased to a much greater degree (Supplemental Figure 6B), suggesting that either pulse-labeled proinsulin normalized to TCA-precipitable cpm underestimates the full magnitude of the proinsulin synthesis defect or that decreased synthesis is only part of the explanation for the decline of proinsulin levels (both points are investigated further below). Indeed, we noted that activation (lipidation) of LC3b to form LC3b-II, suggesting activation of autophagic turnover, was also detectable in young nondiabetic β-Hrd1–KO islets (Figure 2B, lanes 8 and 9).
Figure 5. Steady-state levels of BiP, p58ipk (ERdj6), and phospho-eIF2α in islets of β-Hrd1–KO mice.
(A) Immunoblotting of BiP, p58ipk (ERdj6), and phospho-eIF2α antigens compared with CypB as a loading control. (B) Quantitation of BiP and p58ipk (ERdj6) normalized to CypB (n = 3 per group; mean ± SD; unpaired 2-tailed t test; *P < 0.05); mean random blood glucose for control cohort = 174 mg/dL and for β-Hrd1–KO cohort = 263 mg/dL. Quantitation of phospho-eIF2α normalized to CypB (n = 3 per group; mean ± SD; unpaired 2-tailed t test; *P < 0.05); mean random blood glucose for control cohort = 147 mg/dL and for β-Hrd1–KO cohort = 119 mg/dL.
Acute HRD1 inhibition in pancreatic β cells.
The fact that a rare preweaning β-Hrd1–KO mouse can exhibit nearly normal proinsulin and insulin levels (Figure 2B, lanes 5 and 8) suggested that diminished proinsulin observed in these chronically ERAD-deficient mice represents an adaptation that develops postnatally. To examine acute effects of HRD1 inhibition, we treated INS1E β cells with LS102 for 2 h. LS102 inhibits Hrd1 autoubiquitination (40) and at 10–30 μM also inhibits the ubiquitination of ERAD substrates (41). LS102 was shown in cultured (human alveolar epithelial) cells to trigger significant inhibition of protein (collagen) secretion (42). Acute ERAD inhibition does not significantly change the content of proinsulin in either β cells or their culture medium (Figure 6, A and B). However, based on a recently improved immunoblotting methodology for detection of native and nonnative proinsulin monomers as well as disulfide-linked dimers (43), it was apparent that upon acute HRD1 inhibition, the population of intracellular proinsulin molecules shifts away from native monomers (Figure 6B, lane 2, quantified in Figure 6D), and an increase in phospho-eIF2α (Figure 6B, lane 4, quantified in Figure 6C) accompanies the increase of misfolded proinsulin (Figure 6E). Unlike previous immunoblotting methodology of nonreduced proinsulin that is supersensitive to intermolecular disulfide-linked complexes (Supplemental Figure 7A), the current methodology (43) more accurately detects monomeric proinsulin species (Supplemental Figure 7B). However, regardless of the immunoblotting method, acute HRD1 inhibition results in accumulation of misfolded proinsulin, and similar behavior (diminished native proinsulin) was observed upon LS102 treatment of (nondiabetic) human islets, accompanied by increased phospho-eIF2α and LC3-II (Supplemental Figure 7C and Supplemental Figure 8). Moreover, after an initial 2 h of protein synthesis inhibition in INS1E cells, a cycloheximide washout protocol detected new proinsulin synthesis as an increase of proinsulin abundance (Supplemental Figure 9, lane 7 vs. 6). However, when accompanied by acute HRD1 inhibition, there was little increase in total proinsulin abundance (Supplemental Figure 9, lane 8 vs. 6), but there was increased proinsulin misfolding (Supplemental Figure 9, lane 4 vs. 2; misfolded proinsulin species highlighted with red arrows) and increased phospho-eIF2α (Supplemental Figure 9, lane 8).
Figure 6. Acute treatment of INS1E (β cell line) with LS102.
(A) Intracellular (cells, left) and extracellular (media, right) proinsulin from INS1E cells treated with LS102 (20 μM) for 2 h. (B) Nonreducing SDS-PAGE and immunoblotting of proinsulin from lysates of LS102-treated INS1E cells (lanes 1 and 2, above) or phospho-eIF2α (middle) and LC3b (bottom). (C) Quantitation of phospho-eIF2α (n = 12). (D) Quantitation of native proinsulin as a fraction of total proinsulin (n = 6). (E) Quantitation of nonnative proinsulin dimer (as in B) relative to approximately 6 kDa proinsulin monomer (n = 6). (F) Quantitation of (pre)proinsulin synthesis, as measured in Supplemental Figure 8 (n = 6). (G) Quantitation of LC3-II/LC3-I (n = 12 independent experiments). All quantitation shown is mean ± SD; unpaired 2-tailed t test; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
To test directly whether the increased phospho-eIF2α triggered by acute inhibition of HRD1 activity is linked to a decrease of new proinsulin synthesis, we used 2 independent methods. First, we exploited the recently reported simple method in which a brief combined inhibition of the SEC61 translocon (blocking ER translocation) with simultaneous proteasome inhibition allows detection of new synthesis in the form of (pre)proinsulin (38, 44). Using this method, new (pre)proinsulin synthesis was readily apparent in control INS1E cells (Supplemental Figure 10A, lanes 3 and 5), but upon HRD1 inhibition with LS102 (which triggers an increase of phospho-eIF2α; Figure 6C), (pre)proinsulin synthesis was decreased by approximately 60% (Supplemental Figure 10A, lanes 4 and 6, quantified in Figure 6F). Second, using a conventional 15 min pulse-labeling with 35S–amino acids, upon acute HRD1 inhibition, proinsulin synthesis (normalized to TCA-precipitable cpm) was decreased by approximately 40% (Supplemental Figure 10B). Because the rise in phospho-eIF2α also affects global protein translation (45), proinsulin synthesis normalized against global protein translation (measured by TCA-precipitable cpm) detected a lower fractional inhibition (Supplemental Figure 10B) than observed by normalizing against a stable loading control protein like HSP90 (Figure 6F); nevertheless, both independent methods indicate diminished proinsulin synthesis following acute HRD1 inhibition.
Additionally, among the acute responses to diminished HRD1 activity is LC3b activation (lipidation) to LC3b-II (Figure 6G). Recent findings indicate that the ΣR1 gene product promotes translation of LC3b-I at the ER, and this is the substrate for the generation of lipidated LC3b-II (20). Consistent with these observations, upon siRNA-mediated ΣR1 KD in INS1E β cells (Figure 7A), we observed diminished LC3b-I (Figure 7A, lane 2 vs. 1), and upon acute HRD1 inhibition, we observed diminished LC3b-II (Figure 7A, lane 4 vs. 3, quantified in Figure 7B). INS-832/13 β cells, which grow as relatively flat cells that are favorable for immunofluorescence (46), revealed a primary localization of rodent proinsulin in a juxtanuclear pattern, typical of the normal proinsulin distribution in the Golgi region (25), that was not affected by ΣR1 KD (Figure 7C), strongly suggesting that anterograde trafficking of proinsulin to the Golgi complex continues in ΣR1-deficient β cells. Acute HRD1 inhibition with LS102 caused β cells to lose their strong juxtanuclear proinsulin distribution within 90 min (Figure 7C), implying impaired proinsulin anterograde trafficking with diminished concentration in the Golgi complex. By 2 h of LS1012 treatment, a number of small bright puncta newly appeared that were positive both for proinsulin and FIP200 (Figure 7C), a marker of autophagosome formation (47).
Figure 7. Effects of ΣR1 and LS102 on LC3, and proinsulin distribution in INS1 cells.
(A) Immunoblotting for ΣR1 or LC3 after LS102 treatment of control INS1E cells (scramble oligo) or ΣR1 KD; Hsp90 was a loading control. (B) Quantitation of LC3b-II (normalized to HSP90) in LS102-treated cells after ΣR1 KD (n = 5; unpaired 2-tailed t test; **P < 0.01). (C) INS832-13 cells were transfected with control oligo or ΣR1-KD and after 48 h were treated ± LS102 (20 μM) for up to 2 h (n = 4 per group) before fixation, permeabilization, and immunofluorescence for proinsulin (green) and FIP200, a marker of autophagosome formation (red). Scale bars: 25 μm.
The proinsulin-positive puncta that form upon a 2 h treatment with LS102 were found to be calnexin positive, indicating that they derive from the ER compartment (Supplemental Figure 11, lower left panel), and the formation of such ER-derived proinsulin-positive puncta was blocked in ΣR1-deficient cells (Figure 7C and Supplemental Figure 11, lower right panel), with an intracellular distribution of proinsulin that suggests both failure of proinsulin anterograde trafficking and entry into autophagosomes. Together, the data in Figures 7 and Supplemental Figure 11 support that in β cells, LC3b-I levels (and LC3b-II levels after acute HRD1 inhibition) are dependent upon ΣR1 expression, such that when both ERAD and ER-phagy clearance pathways are acutely inhibited (ERAD by LS102 and autophagy by ΣR1-siRNA), proinsulin becomes entrapped in the ER. Thus, acute HRD1 inhibition in β cells is accompanied by an increase of misfolded proinsulin complexes (Figure 6, B and E, and Supplemental Figures 7 and 8) with decreased native proinsulin (Figure 6D), as well as increased phospho-eIF2α (Figure 6C) that is linked to decreased proinsulin biosynthesis (Supplemental Figure 10), accompanied by autophagy activation (Figure 6G). Autophagy captures some proinsulin (Figure 7C) originating in the ER (Supplemental Figure 11), and the extent of capture is dependent upon expression of ΣR1 (Figure 7, A–C, and Supplemental Figure 11). Indeed, in human islets, aberrant disulfide-linked proinsulin complexes appear destined for a lysosomal fate, as these complexes accumulate upon treatment with lysosomal inhibitors (e.g., chloroquine in Supplemental Figure 12A, quantified in Supplemental Figure 12B and associated with increased LC3-II in Supplemental Figure 12C), events that also occur in β-Hrd1–KO and even WT murine islets (Supplemental Figure 12, D and E). Thus, when β cell HRD1-mediated ERAD is deficient, in addition to islet cell heterogeneity (Figure 3) and ER stress response with increased phospho-eIF2α (Figure 5) accompanied by decreased proinsulin biosynthesis (Supplemental Figure 6A and Supplemental Figure 10), our data suggest that autophagy is also involved in conveying misfolded proinsulin complexes to their ultimate turnover in lysosomes (Supplemental Figure 12).
Discussion
SEL1L-HRD1 (plus associated partner proteins) identify and remove many misfolded proteins from the ER (48). Misfolded proinsulin is subject to ERAD, but the physiological relevance of ERAD capacity in pancreatic β cells has been unclear. What is established is that SEL1L deficiency in β cells causes HRD1 instability (and, thus, hypofunction), and this leads to diminished islet insulin content (17), but the impact on β cell proinsulin content has been controversial (6). HRD1-mediated ERAD capacity for proinsulin might be exceeded in early type 2 diabetes when β cell proinsulin levels — and misfolded proinsulin levels — are unusually elevated, such as has been observed in the islets of young db/db mice (43, 49). Deficient ERAD capacity can be modeled in β cells bearing genetic or pharmacologic deficiency of HRD1. We find that rather than any loss of islet cell mass (17) or size (see Supplemental Figure 2), our findings strongly support the view that deficient β cell ERAD capacity results in a marked decrease of total islet proinsulin as well as insulin (Figure 2, C–E). Nevertheless, based on examination of β-Hrd1–KO mice as well as β cells acutely treated with HRD1 inhibitor, we propose that what is increased is the relative abundance of misfolded proinsulin (Figure 2B, Figure 6, B, D, and E, and Supplemental Figures 7–9), which tends to shift the distribution of intracellular proinsulin away from its normal juxtanuclear Golgi-like concentration with redistribution toward the ER (Figure 3A, Figure 7C, Supplemental Figure 4A, and Supplemental Figure 11).
The loss of β cell ERAD capacity triggers islet cell heterogeneity, including evidence of β cell dedifferentiation with an increase of ALDH3-positive cells (Figure 3 and Supplemental Figure 4B) (17), as has been observed in diabetic states from multiple proximal stresses (including defects that trigger chronically increased cytosolic calcium) — both related and unrelated to ERAD deficiency (17, 26, 27, 50–60). This, as well as the increased abundance of glucagon-positive cells within the islet interior (Figure 3C), may contribute to but may not solely be responsible for the development of diminished islet proinsulin levels, as the increased fraction of misfolded proinsulin in the ER of β cells with either genetically or pharmacologically impaired HRD1 is accompanied by increased phospho-eIF2α (Figure 5 and Figure 7C) that limits proinsulin biosynthesis, which lowers proinsulin levels (61). Although the β-HRD1–KO mice develop diabetes with abnormally small insulin secretory granules (Figures 1 and 4, and Supplemental Figure 5), the increase of phospho-eIF2α (Figure 5, A and B) seems unlikely to simply reflect elevated extracellular glucose (62) because it also occurs upon acute β cell HRD1 inhibition under conditions in which glucose is invariant (Figure 6, B and C, and Supplemental Figure 9), as well as in human islets (Supplemental Figure 8).
ERAD dysfunction can increase ER stress (63), and the increase of phospho-eIF2α is a cardinal feature of the integrated stress response (64) that includes PERK as a potential eIF2α kinase (65). Although we have not proven that PERK is responsible for increased phospho-eIF2α upon HRD1 inhibition, it is certainly a potential candidate, as we have also observed other features typical of ER stress response in the islets of β-Hrd1–KO mice, including a 3- to 6-fold increase of BiP and p58ipk (Figure 5), whereas the increased levels of IRE1α and SEL1L (Supplemental Figure 1C) are more likely to be a direct consequence of diminished ERAD-mediated turnover of these proteins (23). Phospho-eIF2α is a potent suppressor of new proinsulin synthesis (36, 39, 66, 67), which is detected conveniently in a simple and novel assay (Supplemental Figure 10A and Figure 6F) (38, 44). Indeed, upon HRD1 inhibition, the increase of phospho-eIF2α corresponds to a clear block in proinsulin synthesis, and this is confirmed by metabolic labeling studies (Supplemental Figure 10B). Thus, diminished proinsulin synthesis is at least one potentially important contributor to a decrease of intracellular proinsulin levels in β cells with deficient ERAD capacity. The notion that β cell ER stress caused by ERAD deficiency may contribute to impaired β cell Ca2+ homeostasis, resulting in additional β cell defects, is a possibility that also deserves further investigation.
As an additional consideration, we find that acute inhibition of HRD1 function in a β cell line leads to activation (lipidation) of LC3b (Figure 6G), similar to that seen in human islets (Supplemental Figure 8), and upon chronic ERAD inhibition in the islets of β-Hrd1–KO mice (Figure 2B). Trapping autophagy flux in β-Hrd1–KO islets with lysosomal inhibitors — in addition to limiting the turnover of LC3b-II — results in accumulation of proinsulin (Figure 6A) in the form of aberrant disulfide-linked complexes (Figure 6B). The abnormal increase of misfolded proinsulin can be seen within 2 h of HRD1 inhibition; the major misfolded forms to appear are nonnative monomers and disulfide-linked complexes seen in β cell lines and human islets (Figure 7 and Supplemental Figures 7–9). These forms appear preferentially conveyed to lysosomes as they are accumulated by treatment with lysosomal inhibitors (Figure 6). While accumulation of disulfide-linked complexes of proinsulin is also observed upon bafilomycin treatment of islets from control animals (Figure 6A), the data suggest that in a state of diminished ERAD capacity, misfolded proinsulin is susceptible to accumulating in disulfide-linked complexes that can be transported via ER-phagy to lysosomes. Thus, along with suppressed proinsulin synthesis, increased autophagic flux is another key pathway that can account for diminished intracellular proinsulin levels in β cells that have exceeded their HRD1-mediated ERAD capacity.
We have recently reported that when ERAD capacity is exceeded, the IRE1α signaling pathway may also contribute to activation of β cell autophagy (19). Even more recent work indicates that expression of the ΣR1 gene product, which itself has been found to be an ERAD substrate (68), recruits the LC3b mRNA for LC3b-I translation at the ER, which is a precondition that favors its subsequent lipidation to LC3b-II (20). Upon ΣR1 KD in INS1 β cells, acute inhibition of ERAD cannot stimulate the same level of generation of LC3b-II (Figure 7, A and B) and limits the appearance of proinsulin puncta in forming autophagosomes (Figure 7C and Supplemental Figure 11). These findings help to further map the molecular crosstalk between ERAD and autophagic clearance pathways in β cells (17, 19), which in turn impacts proinsulin, and ultimately insulin, levels.
In conclusion, in a state of deficient ERAD capacity, the combined impact of a diminution of mature β cells, an increase in islet β cell phospho-eIF2α with diminished proinsulin synthesis, and enhanced autophagic clearance results in a diminished level of proinsulin that results in markedly decreased insulin content with impaired glucose tolerance (Figure 1, A–C). Indeed, in the setting of diminished insulin storage in β cells (Figure 4), serum insulin cannot keep up with demand (Supplemental Figure 3B), leading to diabetes (Figure 1E). Many additional gene products linked to ER quality control–dependent ubiquitin-proteasomal proteolysis also show genetic evidence of linkage to human type 2 diabetes (such as EDEM3, UFD1, NPL4, BAG1, and FAF1). It is plausible that some of these risk alleles, which act in concert with the SEL1L-HRD1 ERAD machinery, may contribute to a state of diminished ERAD capacity in β cells, potentially leading to versions of the dramatic phenotypes described in the current study.
Methods
Sex as a biological variable.
Animals of both sexes were analyzed and included in the study, as shown in all graphs within the manuscript (males = squares; females = circles); there were similar findings for both sexes. For analyses of glucose tolerance, our study design accounted for sex as a biological variable, as shown in Figure 1. Elsewhere, unless otherwise stated, results from both male and female animals were combined.
Mice.
All mice were in a C57BL6/j background. Hrd1-floxed mice (69) and RIP-Cre mice (21) were used as previously described to generate β cell–specific β-Hrd1–KO mice. Control littermates used for experiments were β cell heterozygous deletion of Hrd1, also known as Synv1. Random blood glucose was measured weekly (and at the time of euthanasia) by a OneTouch Ultra blood glucometer and test strips. For intraperitoneal glucose tolerance tests, mice were fasted for 6 h, d-glucose (1 g/kg body weight) was administered intraperitoneally, and tail vein glucose was monitored (by a OneTouch Ultra glucometer) at different time points thereafter. Where indicated, circulating serum insulin levels were measured using the Mouse Ultrasensitive Insulin ELISA Kit (80-INSMSU-E10; ALPCO). Islet isolation was performed as described previously (49). For overnight recovery, islets were incubated in complete RPMI-1640 medium in a humidified 5% CO2 incubator at 37°C.
Reagents and antibodies.
All reagents and chemicals were from Thermo Fisher Scientific or Sigma-Aldrich, except for the ER translocation inhibitor TL033 (Supplemental Figure 10), which was obtained from T.W. Bell (University of Nevada, Reno, Nevada, USA) (44). SDS-PAGE 4%–12% Bis-Tris or 12% Tris-Glycine NuPage gels were purchased from Thermo Fisher Scientific. Antibodies in this study included mouse mAb antirat proinsulin (CCI-17, Novus, RRID: AB_1107982), guinea pig anti-insulin (Covance, RRID: AB_10013624), rabbit anticyclophilin B (Thermo Fisher Scientific, RRID: AB_2169138), rabbit mAb anti-Hsp90 (Cell Signaling, RRID: AB_2233307), rabbit mAb anti-LC3A/B (Cell Signaling, RRID: AB_2617131), rabbit polyclonal anti-ALDH1A3 (Thermo Fisher Scientific, RRID: AB_2546664), mouse mAb antiglucagon (Abcam, RRID: AB_297642), rabbit polyclonal anticalnexin (Proteintech, RRID: AB_2069033), rabbit anti-IAPP (human residues 25–37, cross-reacting with rodent; BMA Biomedicals, catalog T-4157), rabbit mAb anti-p58ipk (Cell Signaling, RRID: AB_2095213), rabbit mAb anti–phospho-eIF2α (Cell Signaling, RRID: AB_390740), rabbit mAb ΣR1 (Proteintech, RRID: AB_2301712), rabbit polyclonal anti-Hrd1 (Proteintech, RRID: AB_2287023), rabbit polyclonal anti-FIP200 (Proteintech, RRID: AB_10666428), rabbit mAb anti-IRE1α (Cell Signaling, RRID: AB_823545), and mouse mAb antihuman proinsulin Β-C junction sequence (Abmart, RRID: AB_2921300).
Cell culture and ΣR1 KD.
INS1E (obtained from the laboratory of C. Wollheim, University of Geneva, Geneva, Switzerland) and INS/832/13 (obtained from the laboratory of C. Newgard, Duke University, Durham, North Carolina, USA) rat pancreatic β cells were cultured in RPMI-1640 medium (supplemented with 10% FBS, 10 mM HEPES, 1 mM sodium pyruvate, penicillin/streptomycin, and 0.05 mM β-mercaptoethanol). For LS102 treatment, cells were seeded into 12-well plates; and 24 or 48 h later, the cells were fed with fresh complete medium and treated for 2 h with vehicle or 20 μM LS102.
For ΣR1 KD, INS1E (or INS832/13) cells grown to 70% confluence in 6-well plates were transfected with RNAiMax (Invitrogen) containing ΣR1 siRNA (5′-GGCUUGAGCUCACCACCUA annealed with 5′-UAGGUGGUGAGCUCAAGCC) or negative control oligos (Qiagen, 1027281) at a concentration of 100 nM. At 24 h, the cells were fed fresh complete media, and 24 h thereafter, cells were treated with LS102 in fresh media, as described above.
Human islets.
Nondiabetic human pancreatic islets (90% purity, 95% viability; confirmed COVID negative) were generated at Prodo Labs or the University of Michigan Human Islet Core Facility and treated with LS102 as described above: donor 1: 59-year-old female, BMI 28.9 (HbA1c 5.4%), cause of death = stroke; donor 2: 52-year-old female, BMI 28.4 (HbA1c 5.2%), cause of death = anoxic event; donor 3: 38-year-old male, BMI 35.7 (HbA1c 5.6%), cause of death = cardiac event/anoxia; donor 4: 37-year-old male, BMI 35.6 (HbA1c 5.4%), cause of death = anoxia (drug intoxication); donor 5: 66-year-old female, BMI 32.4 (HbA1c 5.5%), cause of death = stroke; donor 6: 68-year-old female, BMI 26.5 (HbA1c 5.3%), cause of death = stroke; donor 7: 62-year-old female, BMI 45.1 (HbA1c 5.4%), cause of death = stroke; donor 8: 69-year-old female, BMI 28.0 (HbA1c 5.5%), cause of death = stroke.
Western blotting.
Cells or islets were lysed in RIPA buffer (25 mM Tris, pH 7.5, 100 nM NaCl, 1% Triton X-100, 0.2% deoxycholic acid, 0.1% SDS, and 10 mM EDTA containing a protease and phosphatase inhibitor cocktail). The collected lysates were spun at 12,000g at 4°C for 15 min, and the supernatant was stored at –80°C. The lysates (5–10 μg) were heated in gel loading buffer (LDS, Invitrogen) at 95°C for 5 min ± 200 mM DTT (for reduced or nonreduced samples, respectively) and resolved by 4%–12% or straight 12% NuPAGE gel. Nonreduced gels were then either untreated or treated with 100 mM DTT at 60°C for 10 min (43) before electrotransfer to nitrocellulose. The membrane was blocked with 5% BSA followed by primary antibody incubation (overnight at 4°C) and then HRP-conjugated secondary antibody incubation at room temperature for 30 min. Blots were developed using Bio-Rad Clarity or Amersham ECL reagent.
Metabolic labeling of INS1E cells and mouse pancreatic islets.
Cells treated ± LS102 for 2 h or isolated from control and HRD1-KO littermates were washed in prewarmed Met/Cys-deficient RPMI medium (Sigma-Aldrich, R7513, ± LS102) and then pulse-labeled with 35S–amino acids (Trans35S label, Revvity Health) for 15 or 20 min at 37°C. The cells or islets were washed with ice-cold PBS containing 20 mM N-ethylmaleimide (NEM)and then lysed in RIPA buffer containing 2 mM NEM and a protease inhibitor cocktail (islets were sonicated in this buffer). Cell lysates (normalized to TCA-precipitable counts) were precleared with pansorbin and immunoprecipitated with anti-insulin antibodies and protein A–agarose (Sigma-Aldrich, 11134515001) overnight at 4°C. Immunoprecipitates were washed and analyzed by reducing 4%–12% gradient NuPAGE or Tris-tricine-urea-SDS-PAGE, fixed, dried, and examined by phosphorimaging. Bands were quantified with ImageJ software.
Immunofluorescence and image analysis.
Paraffin sections of formaldehyde-fixed pancreas were deparaffinized with CitriSolv (Fisher Scientific, 04-355-121) and rehydrated in a decreasing graded series of ethanol followed by heating for antigen retrieval (BioLegend, 927901). Slides were washed with PBS, incubated in TBS blocking buffer containing 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 3 times with TBS/0.1% Tween 20 and mounted with antifade mounting medium-plus-DAPI (Vector Laboratories, H-1800).
INS-832/13 β cells grow as relatively flat cells and thus are favorable for immunofluorescence (46) compared with INS1E cells. INS832/13 cells grown on coverslips (in 12-well plates) ± ΣR1 KD, as described above, were either treated with LS102 or with vehicle (DMSO) for 90 or 120 min beginning at 48 h after siRNA transfection, then briefly washed in PBS, fixed in 4% formaldehyde (20 min at room temperature), washed twice in PBS, permeabilized in 100% cold methanol (10 min at –20°C), washed thrice in TBS, blocked in 3% BSA + 0.2% Tween-20 in TBS (1 h at room temperature), incubated with primary antibody (1:500) in the same blocking buffer (overnight at 4°C), washed thrice again in TBS, incubated with secondary antibody (1:500) in blocking buffer (1 h at room temperature), and finally washed thrice in TBS. Coverslips were placed on a drop of Prolong Gold antifade reagent plus DAPI and mounted on glass slides. For both tissue and cell culture, epifluorescence was imaged on a Nikon A1 confocal microscope, with digital images analyzed by NIS-Elements/ImageJ software.
Transmission electron microscopy.
Islets freshly isolated from control or β-Hrd1–KO mice were fixed in 2.5% glutaraldehyde prepared in 0.1 M sodium cacodylate buffer (pH 7.2), embedded in low-melting-point agarose, cut into approximately 1 mm cubes, postfixed in osmium tetroxide/potassium ferrocyanide, and washed sequentially in sodium cacodylate buffer followed by 0.1 M sodium acetate buffer. The cubes were stained in uranyl acetate (in sodium acetate buffer), dehydrated through a graded ethanol/acetone series, and embedded in Spurr’s resin. Ultrathin sections were examined with a transmission electron microscope (JEOL USA; at 80 kV) and digital camera.
Statistics.
To assess statistical differences between 2 groups, we employed a 2-way ANOVA with Šidák’s multiple-comparison test for data built on 2 independent variables (Figure 1 and Supplemental Figure 3, A and B) or 2-tailed Mann-Whitney U test (Supplemental Figure 1D, Supplemental Figure 2, and Supplemental Figure 5). All other statistical analyses used an unpaired 2-tailed t test for data showing a normal distribution. In each case, the data are presented as the mean ± SD; a P value less than 0.05 was considered statistically significant.
Study approval.
All animal procedures were approved by and done in accordance with the IACUC at the University of Michigan Medical School (PRO00011324).
Data availability.
All data are contained within the figures; additionally, quantitation presented in the figures is documented the Supporting Data Values file. Reagents are available upon request from the corresponding author.
Author contributions
AA, BT, LQ, and PA designed research studies. AA, LH, MA, NFG, EM, SB, JK, CYC, IM, and AH conducted experiments and acquired and analyzed data. DL and DF provided key reagents. PA wrote the manuscript. All authors reviewed, edited, and approved the manuscript.
Funding support
This work is the result of NIH funding and is subject to the NIH Public Access Policy. Through acceptance of this federal funding, the NIH has been given a right to make the work publicly available in PubMed Central.
NIH grant R01DK143292 (to BT, LQ, and PA).
NIH grant R01DK48280 (to PA).
Supplementary Material
Acknowledgments
We thank the morphology and imaging core laboratory affiliated with the University of Michigan Diabetes Research Center (P30DK092926) and the Electron Microscopy Core of the University of Michigan Biomedical Research Core Facilities.
Version 1. 11/18/2025
In-Press Preview
Version 2. 01/16/2026
Electronic publication
Footnotes
Conflict of interest: The authors have declared that no conflict of interest exists.
Copyright: © 2025, Arunagiri et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.
Reference information: J Clin Invest. 2026;136(2):e187341.https://doi.org/10.1172/JCI187341.
Contributor Information
Anoop Arunagiri, Email: ARUNAGIRI@etsu.edu.
Leena Haataja, Email: leenah@med.umich.edu.
Maroof Alam, Email: mdal@umich.edu.
Noah F. Gleason, Email: noahglea@umich.edu.
Emma Mastroianni, Email: emmamast@umich.edu.
Chao-Yin Cheng, Email: chengcha@med.umich.edu.
Jeffrey Knupp, Email: jknupp@umich.edu.
Ibrahim Metawea, Email: Metawea.Ibrahim@mayo.edu.
Anis Hassan, Email: anisf@umich.edu.
Dennis Larkin, Email: dlarkin@umich.edu.
Deyu Fang, Email: fangd@northwestern.edu.
Billy Tsai, Email: btsai@umich.edu.
Ling Qi, Email: xvr2hm@virginia.edu.
Peter Arvan, Email: parvan@umich.edu.
References
- 1.Tiwari A, et al. SDF2L1 interacts with the ER-associated degradation machinery and retards the degradation of mutant proinsulin in pancreatic β-cells. J Cell Sci. 2013;126(pt 9):1962–1968. doi: 10.1242/jcs.117374. [DOI] [PubMed] [Google Scholar]
- 2.He K, et al. PDI reductase acts on Akita mutant proinsulin to initiate retrotranslocation along the Hrd1/Sel1L-p97 axis. Mol Biol Cell. 2015;26(19):3413–3423. doi: 10.1091/mbc.E15-01-0034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cunningham CN, et al. Chaperone-driven degradation of a misfolded proinsulin mutant in parallel with restoration of wild type insulin secretion. Diabetes. 2017;66:741–753. doi: 10.2337/db16-1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Xu B, et al. Estrogens promote misfolded proinsulin degradation to protect insulin production and delay diabetes. Cell Rep. 2018;24(1):181–196. doi: 10.1016/j.celrep.2018.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hoelen H, et al. Proteasomal degradation of proinsulin requires derlin-2, HRD1 and p97. PLoS One. 2015;10(6):e0128206. doi: 10.1371/journal.pone.0128206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hu Y, et al. Endoplasmic reticulum-associated degradation (ERAD) has a critical role in supporting glucose-stimulated insulin secretion in pancreatic β-cells. Diabetes. 2019;68(4):733–746. doi: 10.2337/db18-0624. [DOI] [PubMed] [Google Scholar]
- 7.Xu X, et al. Proteasomal degradation of WT proinsulin in pancreatic beta cells. J Biol Chem. 2022;298(10):102406. doi: 10.1016/j.jbc.2022.102406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hsu HT, et al. Endoplasmic reticulum targeting alters regulation of expression and antigen presentation of proinsulin. J Immunol. 2014;192(11):4957–4966. doi: 10.4049/jimmunol.1300631. [DOI] [PubMed] [Google Scholar]
- 9.Khilji MS, et al. The inducible β5i proteasome subunit contributes to proinsulin degradation in GRP94-deficient β-cells and is overexpressed in type 2 diabetes pancreatic islets. Am J Physiol Endocrinol Metab. 2020;318(6):892–900. doi: 10.1152/ajpendo.00372.2019. [DOI] [PubMed] [Google Scholar]
- 10.Christianson JC, Ye Y. Cleaning up in the endoplasmic reticulum: ubiquitin in charge. Nat Struct Mol Biol. 2014;21(4):325–335. doi: 10.1038/nsmb.2793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lin LL, et al. SEL1L-HRD1 interaction is required to form a functional HRD1 ERAD complex. Nat Commun. 2024;15(1):1440. doi: 10.1038/s41467-024-45633-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ye Y, et al. The AAA ATPase Cdc48/p97 and its partners transport proteins from the ER into the cytosol. Nature. 2001;414(6864):652–656. doi: 10.1038/414652a. [DOI] [PubMed] [Google Scholar]
- 13.Wu T, et al. HRD1, an important player in pancreatic β-cell failure and therapeutic target for type 2 diabetic mice. Diabetes. 2020;69(5):940–953. doi: 10.2337/db19-1060. [DOI] [PubMed] [Google Scholar]
- 14.Iida Y, et al. SEL1L protein critically determines the stability of the HRD1-SEL1L endoplasmic reticulum-associated degradation (ERAD) complex to optimize the degradation kinetics of ERAD substrates. J Biol Chem. 2011;286(19):16929–16939. doi: 10.1074/jbc.M110.215871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sun S, et al. Sel1L is indispensable for mammalian endoplasmic reticulum-associated degradation, endoplasmic reticulum homeostasis, and survival. Proc Natl Acad Sci U S A. 2014;111(5):582–591. doi: 10.1073/pnas.1318114111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Vashistha N, et al. Direct and essential function for Hrd3 in ER-associated degradation. Proc Natl Acad Sci U S A. 2016;113(21):5934–5939. doi: 10.1073/pnas.1603079113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Shrestha N, et al. Sel1L-Hrd1 ER-associated degradation maintains β cell identity via TGF-β signaling. J Clin Invest. 2020;130(7):3499–3510. doi: 10.1172/JCI134874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Son J, Accili D. Reversing pancreatic β-cell dedifferentiation in the treatment of type 2 diabetes. Exp Mol Med. 2023;55(8):1652–1658. doi: 10.1038/s12276-023-01043-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shrestha N, et al. Integration of ER protein quality control mechanisms defines β cell function and ER architecture. J Clin Invest. 2023;133(1):e163584. doi: 10.1172/JCI163584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Knupp J, et al. Sigma-1 receptor recruits LC3 mRNA to ER-associated omegasomes to promote localized LC3 translation enabling functional autophagy. Cell Rep. 2024;43(8):114619. doi: 10.1016/j.celrep.2024.114619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Postic C, et al. Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase. J Biol Chem. 1999;274(1):305–315. doi: 10.1074/jbc.274.1.305. [DOI] [PubMed] [Google Scholar]
- 22.Estall JL, Screaton RA. Of mice and men, redux: modern challenges in β cell gene targeting. Endocrinology. 2020;161(8):bqaa078. doi: 10.1210/endocr/bqaa078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sun S, et al. IRE1α is an endogenous substrate of endoplasmic-reticulum-associated degradation. Nat Cell Biol. 2015;17(12):1546–1555. doi: 10.1038/ncb3266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Orci L, et al. Conversion of proinsulin to insulin occurs coordinately with acidification of maturing secretory vesicles. J Cell Biol. 1986;103(6 pt 1):2273–2281. doi: 10.1083/jcb.103.6.2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Haataja L, et al. Proinsulin intermolecular interactions during secretory trafficking in pancreatic β cells. J Biol Chem. 2013;288(3):1896–1906. doi: 10.1074/jbc.M112.420018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kim-Muller JY, et al. Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic β cells in diabetic mice. Nat Commun. 2016;7:12631. doi: 10.1038/ncomms12631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Son J, et al. Genetic and pharmacologic inhibition of ALDH1A3 as a treatment of β-cell failure. Nat Commun. 2023;14(1):558. doi: 10.1038/s41467-023-36315-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Leenders F, et al. Pancreatic β-cell identity change through the lens of single-cell omics research. Int J Mol Sci. 2024;25(9):4720. doi: 10.3390/ijms25094720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen C-W, et al. Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat Commun. 2022;13(1):4621. doi: 10.1038/s41467-022-32425-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sokolowski EK, et al. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab. 2024;36(11):2468–2488. doi: 10.1016/j.cmet.2024.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Miranda MA, et al. Pancreatic β-cell heterogeneity in health and diabetes: classes, sources, and subtypes. Am J Physiol Endocrinol Metab. 2021;320(4):716–731. doi: 10.1152/ajpendo.00649.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yau B, et al. The role of the beta cell in type 2 diabetes: new findings from the last 5 years. Diabetologia. 2025;68(10):2092–2103. doi: 10.1007/s00125-025-06499-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Brown ME, et al. Pancreatic islet β-cell subtypes are derived from biochemically-distinct and nutritionally-regulated islet progenitors. Nat Commun. 2025;16(1):5758. doi: 10.1038/s41467-025-60831-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Mostafa D, et al. Loss of β-cell identity and diabetic phenotype in mice caused by disruption of CNOT3-dependent mRNA deadenylation. Commun Biol. 2020;3(1):476. doi: 10.1038/s42003-020-01201-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hodish I, et al. In vivo misfolding of proinsulin below the threshold of frank diabetes. Diabetes. 2011;60(8):2092–2101. doi: 10.2337/db10-1671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Harding HP, et al. Diabetes mellitus and exocrine pancreatic dysfunction in perk-/- mice reveals a role for translational control in secretory cell survival. Mol Cell. 2001;7(6):1153–1163. doi: 10.1016/S1097-2765(01)00264-7. [DOI] [PubMed] [Google Scholar]
- 37.Back SH, et al. Translation attenuation through eIF2alpha phosphorylation prevents oxidative stress and maintains the differentiated state in beta cells. Cell Metab. 2009;10(1):13–26. doi: 10.1016/j.cmet.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Xu X, et al. Nutrient-dependent regulation of β-cell proinsulin content. J Biol Chem. 2023;299(7):104836. doi: 10.1016/j.jbc.2023.104836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Harding HP, et al. Uncoupling proteostasis and development in vitro with a small molecule inhibitor of the pancreatic endoplasmic reticulum kinase, PERK. J Biol Chem. 2012;287(53):44338–44344. doi: 10.1074/jbc.M112.428987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yagishita N, et al. RING-finger type E3 ubiquitin ligase inhibitors as novel candidates for the treatment of rheumatoid arthritis. Int J Mol Med. 2012;30(6):1281–1286. doi: 10.3892/ijmm.2012.1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fujita H, et al. The E3 ligase synoviolin controls body weight and mitochondrial biogenesis through negative regulation of PGC-1β. EMBO J. 2015;34(8):1042–1055. doi: 10.15252/embj.201489897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nakajima F, et al. Synoviolin inhibitor LS-102 reduces endoplasmic reticulum stress-induced collagen secretion in an in vitro model of stress-related interstitial pneumonia. Int J Mol Med. 2015;35(1):110–116. doi: 10.3892/ijmm.2014.1984. [DOI] [PubMed] [Google Scholar]
- 43.Arunagiri A, et al. Proinsulin folding and trafficking defects trigger a common pathological disturbance of endoplasmic reticulum homeostasis. Protein Sci. 2024;33(4):e4949. doi: 10.1002/pro.4949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xu X, et al. Role of Sec61α2 translocon in insulin biosynthesis. Diabetes. 2024;73(12):2034–2044. doi: 10.2337/db24-0115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wek RC. Role of eIF2α kinases in translational control and adaptation to cellular stress. Cold Spring Harb Perspect Biol. 2018;10(7):a032870. doi: 10.1101/cshperspect.a032870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Xu X, et al. The role of TRAPγ/SSR3 in preproinsulin translocation into the endoplasmic reticulum. Diabetes. 2022;71(3):440–452. doi: 10.2337/db21-0638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hara T, et al. FIP200, a ULK-interacting protein, is required for autophagosome formation in mammalian cells. J Cell Biol. 2008;181(3):497–510. doi: 10.1083/jcb.200712064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Brodsky JL, et al. Taking out the trash: how misfolded proteins are removed from the endoplasmic reticulum. Fac Rev. 2022;11:29. doi: 10.12703/r-01-0000018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Arunagiri A, et al. Proinsulin misfolding is an early event in the progression to type 2 diabetes. Elife. 2019;8:e44532. doi: 10.7554/eLife.44532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Szabat M, et al. Kinetics and genomic profiling of adult human and mouse β-cell maturation. Islets. 2011;3(4):175–187. doi: 10.4161/isl.3.4.15881. [DOI] [PubMed] [Google Scholar]
- 51.Jonas JC, et al. Chronic hyperglycemia triggers loss of pancreatic beta cell differentiation in an animal model of diabetes. J Biol Chem. 1999;274(20):14112–14121. doi: 10.1074/jbc.274.20.14112. [DOI] [PubMed] [Google Scholar]
- 52.Talchai C, et al. Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure. Cell. 2012;150(6):1223–1234. doi: 10.1016/j.cell.2012.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Farack L, et al. Transcriptional heterogeneity of beta cells in the intact pancreas. Dev Cell. 2019;48(1):115–125. doi: 10.1016/j.devcel.2018.11.001. [DOI] [PubMed] [Google Scholar]
- 54.van der Meulen T, et al. Virgin beta cells persist throughout life at a neogenic niche within pancreatic islets. Cell Metab. 2017;25(4):911–926. doi: 10.1016/j.cmet.2017.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Brereton MF, et al. Reversible changes in pancreatic islet structure and function produced by elevated blood glucose. Nat Commun. 2014;5:4639. doi: 10.1038/ncomms5639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Salinno C, et al. CD81 marks immature and dedifferentiated pancreatic β-cells. Mol Metab. 2021;49:101188. doi: 10.1016/j.molmet.2021.101188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Motomura K, et al. Single-cell transcriptome profiling of pancreatic islets from early diabetic mice identifies Anxa10 for Ca2+ allostasis toward β-cell failure. Diabetes. 2024;73(1):75–92. doi: 10.2337/db23-0212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Alam M, et al. Predisposition to proinsulin misfolding as a genetic risk to diet-induced diabetes. Diabetes. 2021;70(11):2580–2594. doi: 10.2337/db21-0422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Barbetti F, et al. Hyperglucagonemia in an animal model of insulin- deficient diabetes: what therapy can improve it? Clin Diabetes Endocrinol. 2016;2:11. doi: 10.1186/s40842-016-0029-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Stancill JS, et al. Chronic β-cell depolarization impairs β-cell identity by disrupting a network of Ca2+-regulated genes. Diabetes. 2017;66(8):2175–2187. doi: 10.2337/db16-1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zavarzadeh PG, et al. Exploring proinsulin proteostasis: insights into beta cell health and diabetes. Front Mol Biosci. 2025;12:1554717. doi: 10.3389/fmolb.2025.1554717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Elouil H, et al. Acute nutrient regulation of the unfolded protein response and integrated stress response in cultured rat pancreatic islets. Diabetologia. 2007;50(7):1442–1452. doi: 10.1007/s00125-007-0674-4. [DOI] [PubMed] [Google Scholar]
- 63.Sim HJ, et al. Augmented ERAD (ER-associated degradation) activity in chondrocytes is necessary for cartilage development and maintenance. Sci Adv. 2022;8(3):eabl4222. doi: 10.1126/sciadv.abl4222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Harding HP, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003;11(3):619–633. doi: 10.1016/S1097-2765(03)00105-9. [DOI] [PubMed] [Google Scholar]
- 65.Harding HP, et al. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 1999;397(6716):271–274. doi: 10.1038/16729. [DOI] [PubMed] [Google Scholar]
- 66.Scheuner D, et al. Control of mRNA translation preserves endoplasmic reticulum function in beta cells and maintains glucose homeostasis. Nat Med. 2005;11(7):757–764. doi: 10.1038/nm1259. [DOI] [PubMed] [Google Scholar]
- 67.Liu M, et al. Proinsulin disulfide maturation and misfolding in the endoplasmic reticulum. J Biol Chem. 2005;280(14):13209–13212. doi: 10.1074/jbc.C400475200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhou Z, et al. Endoplasmic reticulum-associated degradation regulates mitochondrial dynamics in brown adipocytes. Science. 2020;368(6486):54–60. doi: 10.1126/science.aay2494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yang H, et al. Hrd1-mediated BLIMP-1 ubiquitination promotes dendritic cell MHCII expression for CD4 T cell priming during inflammation. J Exp Med. 2014;211(12):2467–2479. doi: 10.1084/jem.20140283. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data are contained within the figures; additionally, quantitation presented in the figures is documented the Supporting Data Values file. Reagents are available upon request from the corresponding author.








