International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Diabetes is one of the most common and serious global health problems, as well as one of fastest growing chronic diseases (1). For the 10th edition of the IDF Diabetes Atlas, by 2045, the global prevalence of diabetes among adults aged 20-79 years is projected to increase from 9% (463 million adults) in 2019 to 12.2% (783.2 million) (2). Among all cases of diabetes, type 2 diabetes mellitus (T2DM) accounts for ~90% of all cases (3). Obesity is an important risk factor for T2D (4). The increase in free fatty acids (FFAs) caused by obesity plays a crucial role in the occurrence and development of T2D (4).
T2D is characterized by insufficient insulin secretion and chronic hyperglycemia caused by pancreatic β-cell dysfunction (5). Regulating glucose transporter 2 (GLUT2) is a key protein for β-cell function and a transmembrane protein expressed in pancreatic β-cells, liver, kidneys and intestines, playing a key role in glucose sensing and insulin secretion (6). GLUT2 has a low affinity but high transport capacity for glucose, allowing β-cells to rapidly take up glucose when blood levels rise. This triggers metabolic pathways that increase intracellular ATP, leading to insulin secretion (7). The dysregulation of GLUT2 expression or function impairs glucose sensing and insulin release, which has been reported to contribute to β-cell dysfunction and the pathogenesis of T2D (8,9).
Among the various factors implicated in β-cell dysfunction, chronic exposure to high levels of FFAs is a major factor (10), with evidence suggesting that elevated FFAs have deleterious effects on β-cell function and survival, a phenomenon often referred to as lipotoxicity (5,11). Research has shown that metabolic stressors, such as elevated FFAs, can alter GLUT2 expression, further exacerbating β-cell dysfunction (12). A saturated fatty acid, palmitic acid (PA), has been shown to impair glucose-stimulated insulin release (13-17), and can induce lipotoxicity, leading to β-cell dysfunction and apoptosis (often referred to as lipotoxic β-cell apoptosis) (10,15,18,19).
PA can exert these detrimental effects on β-cells through various mechanisms, including the induction of endoplasmic reticulum (ER) stress, oxidative stress and inflammatory response (20). Given their critical role in insulin synthesis, β-cells have a highly developed ER network to synthesize insulin. Consequently, ER stress is particularly important in β-cell dysfunction (21). Under stress conditions, it leads to the accumulation of misfolded proteins in the ER and the activation of the unfolded protein response (UPR) (22). Long-term or excessive ER stress eventually triggers β-cell apoptosis, leading to the gradual loss of functional β-cells in patients with T2D (23).
A recent study highlighted the role of specific ER proteins in regulating ER stress and β-cell survival (24). Endoplasmic reticulum-resident protein 46 (ERp46), a thiol-disulfide oxidoreductase that is highly expressed in endothelial cells, pancreatic β-cells, hepatocytes and hypoxic tissues (25-27), plays a crucial role in protein folding and redox regulation (24,28). ERp46 has been identified as a key regulator of cellular homeostasis, and its activity is particularly important under conditions of ER stress, including maintenance of β-cell function and involvement in insulin secretion (29). In addition, GLUT2 plays a role in the intracellular signaling pathways involved in glucose uptake and metabolism in ER stress and apoptosis (30), suggesting that ERp46 may also be linked to these processes, although this has not been directly demonstrated. Despite the potential importance of ERp46 in β-cell function and the fact that its expression is reduced in β-cells in diabetic mouse models and under high glucose stimulation (31), the mechanisms through which ERp46 regulates β-cells in diabetic mouse models and, particularly how it may link ER stress to GLUT2 expression, remain unclear. The aim of the present study was to explore the role of ERp46 in pancreatic β-cells, particularly in the regulation of insulin secretion under PA-induced lipotoxic stress. By elucidating the molecular mechanism through which ERp46 affects β-cell function, this study may help develop novel therapeutic strategies targeting ER stress to preserve β-cell function in patients with T2D.
The β-TC6 mouse insulinoma cell line was purchased from the National Collection of Authenticated Cell Cultures. Cells were maintained in DMEM (Sigma-Aldrich; Merck KGaA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) at 37˚C and 5% CO2. A stock solution of PA (Sigma-Aldrich; Merck KGaA) was prepared in 100% ethanol at 55˚C until it was completely dissolved at a concentration of 100 mM. The final ethanol concentration in culture did not exceed 0.1% (v/v), and vehicle controls were included accordingly. Prior to PA treatment, the stock solution was added to DMEM with 10% FBS at the indicated concentrations.
SC79 (Selleck Chemicals) was dissolved in DMSO to prepare a 50-mM stock solution stored at -20˚C. For experiments, the stock was diluted in DMEM to a final concentration of 5 or 10 µM, ensuring the DMSO concentration did not exceed 0.1%. β-TC6 cells at 70-80% confluency were treated with SC79 for 4 h at 37˚C, 5% CO2. DMSO-treated cells were used as controls. Following treatment, cells were harvested for protein extraction or insulin secretion assays.
RNA-seq data (GSE53949) was downloaded from the GEO database (32) and analyzed in RStudio (version 1.4.1717; Posit PBC) using the DESeq2 package (version 1.30.1; https://bioconductor.org/packages/release/bioc/html/DESeq2.html). GSE53949 included five control and five PA-treated human pancreatic islets samples; all ten samples were used for differential and correlation analyses. Following initial quality control, normalization was performed to adjust for library size variations. Differential expression analysis identified genes with |log2FC|>1 and P<0.05 as significant, and P-values were adjusted using the Benjamini-Hochberg method to control for false discovery rate. Volcano plots were generated with ggplot2 (version 3.3.5; http://ggplot2.tidyverse.org), where upregulated genes were highlighted in red and downregulated genes in green. Functional enrichment, including Gene Ontology (http://geneontology.org) and Kyoto Encyclopedia of genes and genomes (KEGG; https://www.genome.jp/kegg/) pathway analysis, was conducted using the clusterProfiler package (version 4.0.5; http://bioconductor.org/packages/release/bioc/html/clusterProfiler.html), and results were visualized through bar plots. To assess the relationship between thioredoxin domain-containing protein 5 [TXNDC5 (ERp46)] and solute carrier family 2 member 2 [SLC2A2 (GLUT2)], Pearson's correlation analysis was conducted using normalized RNA-seq expression data. A linear regression model was applied, and the coefficient of determination (R²) was calculated to quantify the strength of the correlation. Scatter plots with fitted regression lines were generated to visualize the results.
Total RNA was extracted from β-TC6 cells using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). First-strand complementary DNA was synthesized using the Hifair® II 1st Strand cDNA Synthesis Kit (Shanghai Yeasen Biotechnology Co., Ltd.), according to the manufacturer's instructions. Subsequently, RT-qPCR was performed using the Hieff® qPCR SYBR Green Master Mix (Shanghai Yeasen Biotechnology Co., Ltd.) according to the standard method. The thermocycling conditions were as follows: Initial denaturation at 95˚C for 2 min, followed by 40 cycles of denaturation at 95˚C for 10 sec and annealing/extension at 60˚C for 30 sec. A melting curve analysis was performed to verify amplification specificity. The relative expression (fold) was calculated using the comparative method 2-ΔΔCq method (33). Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used for normalization. The primer sequences are shown in Table I.
The culture dish in which β-TC6 cells were seeded was washed twice with cold PBS to completely remove the culture medium, and the cells were lysed using RIPA lysis buffer (Beijing Solarbio Science & Technology Co., Ltd.) supplemented with protease inhibitors. After gently scraping the cells with a cell scraper, they were transferred to a 1.5 ml Eppendorf tube and incubated on ice for 30 min for lysis. The cells were vortexed every 5 min to enhance protein extraction. The lysate was centrifuged at 12,000 x g for 15 min at 4˚C to precipitate cell debris. The supernatant containing the extracted protein was carefully transferred to a new tube for subsequent analysis.
Protein concentration was determined using the bicinchoninic acid (BCA) assay (Thermo Fisher Scientific, Inc.). A standard curve was prepared using bovine serum albumin (Thermo Fisher Scientific, Inc.) as a standard. Protein samples were added to different wells of a 96-well plate. Subsequently, 200 µl of BCA reagent mixture (comprised of reagent A and reagent B mixed at a ratio of 50:1) was added to each well. The plate was incubated at 37˚C for 30 min, and absorbance was measured at 562 nm using a microplate reader. Protein concentration was calculated by comparing the absorbance of the sample with the standard curve. Depending on the concentration, protein samples were diluted to 1 µg/µl using RIPA and then added to protein loading buffer, boiled at 100˚C for 5 min, aliquoted and stored in a freezer at -80˚C.
Protein samples (50 µg) were separated by 10% SDS-PAGE and transferred to PVDF membranes (Wuhan Servicebio Technology Co., Ltd.). Following blocking with 5% skim milk in TBST for 1 h at room temperature, the membranes were incubated with primary antibodies overnight at 4˚C: Anti-ERp46 (1:1,000; cat. no. sc-271667; Santa Cruz Biotechnology, Inc.), anti-GLUT2 (1:1,000; cat. no. K006592P; Beijing Solarbio Science & Technology Co., Ltd.), anti-p-Akt (1:1,000; cat. no. GB150002), anti-Akt (1:1,000; cat. no. GB111114), anti-pancreatic and duodenal homeobox 1 (PDX1; 1:1,000; cat. no. GB11917), and anti-tubulin (1:2,000; cat. no. GB11017; all from Wuhan Servicebio Technology Co., Ltd.), anti-binding immunoglobulin protein (BiP; 1:1,000; cat. no. 3177T), C/EBP homologous protein (CHOP; 1:1,000; cat. no. 2895T), and anti-Na+/K+-ATPase [1:1,000; cat. no. 3010S; all from Cell Signaling Technology, Inc.]. Following washing three times with TBST (TBS containing 0.1% Tween-20) for 10 min each time, the membranes were incubated with secondary antibodies: Anti-rabbit IgG, HRP-linked antibody (cat. no. 7074) and anti-mouse IgG, HRP-linked antibody (cat. no. 7076) (both 1:5,000; Cell Signaling Technology, Inc.), for 1 h at room temperature. Protein bands were visualized using enhanced chemiluminescence (Thermo Fisher Scientific, Inc.), and band intensities were quantified using ImageJ software (version 1.53t; National Institutes of Health).
Supernatants of β-TC6 cells exposed to various concentrations (0, 0.1, 0.25 and 0.5 mM) of PA for 24 h at 37˚C were collected, and insulin levels were measured using a mouse insulin ELISA kit [cat. no. 634-01481 (formerly AKRIN-011T) FUJIFILM Wako Pure Chemical Corporation], according to the manufacturer's instructions. Absorbance was measured at 450 nm, and insulin concentrations were calculated using a standard curve.
For the knockdown of ERp46, 1x106 cells were seeded into a 6-well plate. A total of 50 pmol ERp46 small interfering (siRNA) (sense, 5'-GUACUCGGUACGAGGUUAUTT-3' and antisense, 5'-AUAACCUCGUACCGAGUACTT-3') or negative control siRNA (sense, 5'-UUCUCCGAACGUGUCACGUTT-3' and antisense, 5'-ACGUGACACGUUCGGAGAATT-3') were transfected using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions (at 3˚C for 6 h; typically 5 µl reagent per well in Opti-MEM). The transfection medium was then replaced with DMEM containing 12% FBS, and cells were cultured for 24 h at 37˚C. The expression of ERp46 was detected using western blotting and RT-qPCR.
Statistical analysis was performed using GraphPad Prism 8.0 (GraphPad Software, Inc.). Data are presented as the mean ± standard error of the mean from three to five independent experiments, with each experiment including n=6 biological replicates per group. Statistical comparisons between two groups were performed using an unpaired two-tailed Student's t-test, and multiple-group comparisons were analyzed using ANOVA followed by Sidak's multiple-comparisons test. P<0.05 was considered to indicate a statistically significant difference.
As aforementioned, FFAs can cause lipotoxicity in pancreatic β-cells (10,15,19), and it was found that following PA stimulation, pancreatic β-cells secreted less insulin, which was proportionate to the dose of PA (Fig. 1A), consistent with previous studies (16,17). When stimulated with 0.5 µM PA, insulin secretion decreased over time; however, there was no significant difference between 24 and 48 h, suggesting that the effect reached saturation (Fig. 1A). To further investigate the cause of the reduced insulin secretion, RNA-seq analysis of a public dataset (GSE53949) was performed. By analyzing the RNA-seq results of PA-stimulated β-cells compared with the control group, TXNDC5 (gene name of ERp46) was significantly upregulated (Fig. 1B). This upregulation was supported by enrichment analysis, which highlighted the activation of ER stress-related pathways in β-cells following PA stimulation (Fig. 1C). These findings were consistent with previous reports, in which the upregulation of TXNDC5 was associated with its role in promoting correct protein folding and maintaining redox homeostasis under ER stress conditions (25-27).
By contrast, the key glucose transporter SLC2A2 (gene name of GLUT2) in β-cells was significantly downregulated following PA stimulation. Enrichment analysis identified the ‘negative regulation of transport’ pathway (Fig. 1D), highlighting the importance of GLUT2 in β-cell function. The downregulation of GLUT2 suggested a reduced glucose-transport capacity and was compatible with the decreased insulin secretion observed in Fig. 1A. Previous studies have demonstrated that GLUT2 plays a critical role in glucose sensing and insulin secretion in β-cells (8,9), although the present data did not directly establish causality. Consistently, RT-qPCR confirmed that SLC2A2 mRNA was decreased while TXNDC5 mRNA was increased in PA-treated cells (Fig. 1E). Western blotting further demonstrated decreased GLUT2 protein and increased ERp46 protein levels following PA treatment (Fig. 1F). In conclusion, PA stimulation was associated with the activation of ER stress-related pathways and reduced GLUT2 expression and insulin secretion in β-cells.
ERp46 knockdown efficiency was first verified. Western blotting confirmed a robust reduction of ERp46 protein following siERp46 transfection (Fig. 2A). This result validated the effectiveness of the knockdown system for subsequent experiments. Next, GLUT2 protein was examined across the four conditions (Control, PA, siERp46 and PA + siERp46). As shown in Fig. 2B, PA alone decreased GLUT2, whereas siERp46 alone had no detectable effect. Of note, the combination of siERp46 with PA further reduced GLUT2 compared with PA alone. These findings indicated that ERp46 is not essential for basal GLUT2 expression, but contributes to the maintenance of GLUT2 under lipotoxic stress.
Functionally, insulin secretion followed the same pattern (Fig. 2C): PA reduced insulin release; siERp46 alone did not alter secretion; and siERp46 plus PA led to a further decrease relative to PA alone. This indicates that the protective role of ERp46 becomes evident only in the presence of PA-induced stress. Since ERp46 is an ER oxidoreductase, ER stress markers were assessed. PA increased BiP and CHOP, and siERp46 further increased their levels under PA, whereas siERp46 alone showed no significant change (Fig. 2D). These results are consistent with the interpretation that ERp46 depletion aggravates ER stress in β-cells exposed to PA. PDX1, a β-cell identity/functional factor (12,30), was also evaluated. PA decreased PDX1 mRNA and protein levels, while siERp46 did not produce an additional significant reduction beyond PA in our conditions (Fig. 2E and F). This suggested that ERp46 depletion primarily impacted GLUT2 and insulin secretion rather than β-cell identity markers under PA stress.
Collectively, these data indicated that ERp46 depletion exacerbates PA-induced ER stress and is associated with a greater loss of GLUT2 and insulin secretion, whereas ERp46 knockdown alone has minimal effects at baseline. Thus, ERp46 appears to play a stress-dependent protective role in sustaining GLUT2 expression and β-cell functionality.
The relationship between TXNDC5 (ERp46) and SLC2A2 (GLUT2) was first examined using the public RNA-seq dataset (GSE53949). In the correlation plot, each dot represents one RNA-seq sample from GSE53949 (PA-stimulated or control β-cell samples). A modest positive correlation was observed (R²=0.4001, P=0.0497; Fig. 3A), supporting an association between ERp46 and GLUT2 under lipotoxic conditions. Next, AKT activation was assessed. Total AKT (t-AKT) remained unchanged across groups, whereas PA decreased phosphorylated AKT (p-AKT). Of note, siERp46 alone had little effect, but siERp46 in the presence of PA further reduced p-AKT (Fig. 3B). These findings indicated that ERp46 depletion exacerbated the PA-induced suppression of AKT phosphorylation, while having a minimal impact under basal conditions.
To determine whether AKT activation is sufficient to counteract the PA/siERp46 effects, cells were treated with the AKT activator SC79. SC79 increased p-AKT and partially restored GLUT2 protein in PA-exposed cells under both control siRNA and siERp46 conditions (Fig. 3C). Functionally, SC79 also improved insulin secretion that was reduced by PA and further diminished by siERp46 (Fig. 3D). To evaluate specificity, an additional membrane protein and ER-stress markers were probed. Na+/K+-ATPase was selected as a representative plasma membrane housekeeping protein whose expression remains stable under metabolic or ER stress conditions, serving as a control for nonspecific changes in membrane protein abundance. Na+/K+-ATPase levels were unchanged across treatments, and SC79 did not reduce PA-induced increases of BiP or CHOP (Fig. 3E), indicating that AKT activation does not broadly elevate membrane proteins or alleviate ER stress but can still restore GLUT2 and insulin secretion.
In conclusion, ERp46 depletion was demonstrated to enhance the PA-induced loss of p-AKT, GLUT2 and insulin secretion, while pharmacological AKT activation rescued GLUT2 and insulin without reducing ER-stress markers. These findings supported a model in which AKT acts downstream or in parallel to ER-stress pathways, with ERp46 helping to preserve AKT activity and β-cell function specifically under lipotoxic stress.
The present study revealed the critical role of ERp46 in regulating GLUT2 expression and insulin secretion in pancreatic β-cells under PA-induced lipotoxic stress. These findings demonstrated that ERp46 exerted a protective effect on β-cell function, primarily by alleviating PA-induced ER stress, which in turn helps sustain AKT phosphorylation and maintain GLUT2 expression. These observations suggested that AKT may act downstream or in parallel to ER stress, rather than being directly regulated by ERp46 under basal conditions. However, the possibility that ERp46 may influence AKT signaling more directly through effects on the folding or stability of upstream signaling components cannot be excluded and should be further investigated. The findings of the present study provide novel insights into the molecular mechanisms underlying β-cell dysfunction in T2DM.
PA-induced ER stress is a well-known contributor to β-cell dysfunction, leading to impaired insulin secretion and increased cell death through mechanisms such as protein misfolding and oxidative stress (23,30). Consistent with these observations, the present study showed that PA stimulation significantly decreased GLUT2 expression and insulin secretion in β-cells. Previous studies have shown that ER stress disrupts glucose sensing by reducing GLUT2 expression and impairing insulin granule exocytosis (22,23). Furthermore, prolonged ER stress was shown to contribute to β-cell apoptosis, further limiting insulin secretion capacity (10,18). The chronic activation of the UPR has been demonstrated to further promote β-cell apoptosis and functional decline under lipotoxicity (10,34). Of note, ERp46 expression was upregulated in response to PA, suggesting a compensatory mechanism to counteract ER stress. However, the knockdown of ERp46 further reduced GLUT2 expression and exacerbated the decline in insulin secretion, highlighting its protective role in maintaining β-cell functionality. As a member of the protein disulfide isomerase (PDI) family, ERp46 is essential for protein folding and redox homeostasis in the ER (6,12). This is consistent with its reported ability to mitigate cellular stress by ensuring proteostasis under adverse conditions. Concordantly, multiple PDI paralogs (such as PDI family A member 1 (PDIA1)/pyruvate dehydrogenase E1 component subunit beta and ERp57/PDIA3 modulate β-cell stress adaptation and insulin biosynthesis, underscoring a conserved ER proteostasis axis in the endocrine pancreas (24,35). Previous studies have further emphasized the role of ERp46 in alleviating ER stress and apoptosis in β-cells under diabetic conditions (22,31), suggesting its broader function in metabolic regulation. A recent comprehensive review of TXNDC5/ERp46 across diseases also highlighted its metabolic relevance and therapeutic tractability (29).
In addition to its role in maintaining ER homeostasis, ERp46 may influence GLUT2 expression, at least partly through the modulation of the AKT signaling pathway. AKT activation has been widely reported as a crucial regulator of glucose uptake and insulin secretion in β-cells (30,36). In the present study, PA stimulation significantly reduced p-AKT, and this effect was further exacerbated by ERp46 knockdown, suggesting that ERp46 supports AKT activation under lipotoxic conditions, which in turn may help preserve GLUT2 expression. Although total AKT levels remained unchanged across experimental groups, PA stimulation significantly reduced p-AKT levels, and this reduction was further exacerbated by ERp46 knockdown. This suggested that ERp46 primarily sustains AKT phosphorylation indirectly by alleviating ER stress, as siERp46 had no effect under baseline conditions but markedly suppressed AKT activity in the presence of PA. Thus, AKT is more likely to act downstream or in parallel to ER stress rather than being directly controlled by ERp46. Of note, treatment with SC79, a direct AKT activator, successfully rescued GLUT2 expression and restored insulin secretion in ERp46-depleted cells. This is consistent with broader evidence that phosphoinositide 3-kinase (PI3K)/AKT signaling is a central survival and metabolic pathway in β-cells and a candidate lever to counter lipotoxic dysfunction (37,38). These findings provided direct evidence that ERp46 supports GLUT2 expression and β-cell functionality by modulating AKT activation.
UPR signaling has been reported to intersect with AKT activity in other cellular contexts. For example, PERK-eIF2α signaling can suppress insulin biosynthesis, whereas adaptive inositol-requiring enzyme 1-X-box binding protein 1 and activating transcription factor 6 branches promote β-cell survival and proteostasis (39,40). Additional studies have suggested that maladaptive ER stress can impair AKT phosphorylation, while protective UPR responses may sustain it (41,42). Although these findings indicated potential crosstalk between UPR and AKT, whether this mechanism operates in β-cells under lipotoxic stress remains uncertain. The present data that ERp46 depletion reduces p-AKT under PA stress raise the possibility that ERp46 may act by stabilizing ER proteostasis, thereby preventing maladaptive UPR activation and indirectly supporting AKT activity. Nevertheless, the alternative hypothesis that ERp46 may also regulate AKT through more direct mechanisms, cannot be excluded.
The positive correlation between ERp46 and GLUT2 highlights the importance of maintaining ER homeostasis in preserving β-cell function under lipotoxic stress. Furthermore, the ability of AKT activation to mitigate the negative effects of ERp46 depletion suggests that targeting the ERp46-AKT-GLUT2 axis could offer a therapeutic strategy for improving β-cell survival and insulin secretion in T2DM. In parallel, interventions that lower ER stress, such as chemical chaperones or UPR modulators, have demonstrated the protection of β-cell mass and function in preclinical models and early translational studies (43-46), supporting an ‘ER-proteostasis-first’ approach that aligned with the present model. These findings supported the rationale that enhancing ERp46 activity may represent a novel strategy to stabilize β-cell proteostasis and insulin secretory function.
Beyond diabetes, ER stress is a convergent mechanism in multiple chronic diseases, including cardiovascular disease, neurodegeneration and cancer (40,47-50). Placing these results in this broader context underscores the translational significance of modulating ER proteostasis; targeting ERp46 or allied PDI nodes could complement metabolic therapies and potentially benefit co-morbid conditions characterized by secretory stress. Of note, the spleen has recently been recognized as an active immunometabolic hub that communicates with the gut and liver to shape lipid and glucose metabolism, as well as systemic inflammation, forming distinct spleen-organ axes (51). In addition, in a high-fat/streptozotocin rat model with splenectomy, adipose tissue-derived stem cells protected against T2D through the induction of spleen-derived IL-10; this benefit was blunted by splenectomy, highlighting a spleen-IL-10 anti-inflammatory circuit with a metabolic impact (52). An independent study further showed that spleen-derived or exogenous IL-10 can dampen obesity-associated inflammation and insulin resistance in liver and adipose tissues (53). Of note, IL-10 has also been demonstrated to alleviate ER stress and apoptosis in non-islet tissues, such as cardiomyocytes under doxorubicin challenge (54) and skeletal muscle of aged mice (55). These findings suggested that splenic IL-10-driven anti-inflammatory circuits could, in principle, buffer β-cell ER stress and indirectly preserve AKT phosphorylation and GLUT2 expression, consistent with our ERp46-proteostasis model.
Despite these insights, the precise mechanisms through which ERp46 regulates AKT activation remain to be fully elucidated. It is possible that ERp46 interacts directly with components of the PI3K/AKT pathway, but it is also plausible that the effects are mediated indirectly through ER stress and UPR signaling. Further studies are warranted to dissect these molecular interactions and to explore the in vivo role of ERp46, particularly in diabetic animal models. In addition, complementary approaches such as conditional β-cell-specific ERp46-knockout models or the pharmacological modulation of ER stress may provide more definitive evidence. In addition, the involvement of other ER stress-related pathways in regulating GLUT2 expression and insulin secretion deserves further investigation. Collectively, these findings not only enhanced our understanding of β-cell dysfunction under lipotoxic stress but also underscored the potential of ERp46 as a promising therapeutic target for diabetes and possibly other ER stress-related diseases.
In conclusion, the present study demonstrated that ERp46 is a key regulator of GLUT2 expression and insulin secretion in β-cells under lipotoxic stress. Rather than directly activating AKT, ERp46 primarily alleviated PA-induced ER stress, which in turn supported AKT phosphorylation and helped preserve GLUT2 expression. These findings provided a mechanistic basis for the development of therapeutic strategies targeting ER stress modulators in combination with ERp46- and AKT-related pathways to combat β-cell dysfunction in T2DM.
The authors would like to thank Zhongshan Hospital of Xiamen University (Xiamen, China) for providing laboratory facilities and technical support.
Funding: The present study was supported by the Guidance Project of Xiamen Science and Technology Bureau (grant no. 3502Z20189038).
The data generated in the present study may be requested from the corresponding author.
DC acquired the funding and contributed to the conceptualization, data curation, formal analysis, investigation, methodology, and drafting and revision of the manuscript. CH contributed to the conceptualization, investigation, methodology, project administration, supervision and manuscript revision. XC contributed to the investigation, data curation and formal analysis. YT and KW contributed to data curation and formal analysis. DC and CH confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Heald AH, Stedman M, Davies M, Livingston M, Alshames R, Lunt M, Rayman G and Gadsby R: Estimating life years lost to diabetes: Outcomes from analysis of National diabetes audit and office of National statistics data. Cardiovasc Endocrinol Metab. 9:183–185. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, Stein C, Basit A, Chan JCN, Mbanya JC, et al: IDF diabetes atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 183(109119)2022.PubMed/NCBI View Article : Google Scholar | |
|
Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, et al: Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International diabetes federation diabetes atlas, 9th edition. Diabetes Res Clin Pract. 157(107843)2019.PubMed/NCBI View Article : Google Scholar | |
|
Piché ME, Tchernof A and Després JP: Obesity phenotypes, diabetes, and cardiovascular diseases. Circ Res. 126:1477–1500. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Eizirik DL, Pasquali L and Cnop M: Pancreatic β-cells in type 1 and 2 diabetes mellitus: Different pathways to failure. Nat Rev Endocrinol. 16:349–362. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Thorens B: GLUT2, glucose sensing and glucose homeostasis. Diabetologia. 58:221–232. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Mueckler M and Thorens B: The SLC2 (GLUT) family of membrane transporters. Mol Aspects Med. 34:121–138. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Thorens B, Guillam MT, Beermann F, Burcelin R and Jaquet M: Transgenic reexpression of GLUT1 or GLUT2 in pancreatic beta cells rescues GLUT2-null mice from early death and restores normal glucose-stimulated insulin secretion. J Biol Chem. 275:23751–23758. 2000.PubMed/NCBI View Article : Google Scholar | |
|
Guillam MT, Dupraz P and Thorens B: Glucose uptake, utilization, and signaling in GLUT2-null islets. Diabetes. 49:1485–1491. 2000.PubMed/NCBI View Article : Google Scholar | |
|
Lytrivi M, Castell AL, Poitout V and Cnop M: Recent insights into mechanisms of β-cell lipo- and glucolipotoxicity in type 2 diabetes. J Mol Biol. 432:1514–1534. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Prentki M, Peyot ML, Masiello P and Madiraju SRM: Nutrient-induced metabolic stress, adaptation, detoxification, and toxicity in the pancreatic β-cell. Diabetes. 69:279–290. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Ashcroft FM and Rorsman P: Diabetes mellitus and the β cell: The last ten years. Cell. 148:1160–1171. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Xie T, So WY, Li XY and Leung PS: Fibroblast growth factor 21 protects against lipotoxicity-induced pancreatic β-cell dysfunction via regulation of AMPK signaling and lipid metabolism. Clin Sci (Lond. 133:2029–2044. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Biden TJ, Robinson D, Cordery D, Hughes WE and Busch AK: Chronic effects of fatty acids on pancreatic beta-cell function: New insights from functional genomics. Diabetes. 53 (Suppl 1):S159–S165. 2004.PubMed/NCBI View Article : Google Scholar | |
|
Cnop M: Fatty acids and glucolipotoxicity in the pathogenesis of type 2 diabetes. Biochem Soc Trans. 36:348–352. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Barlow J, Jensen VH, Jastroch M and Affourtit C: Palmitate-induced impairment of glucose-stimulated insulin secretion precedes mitochondrial dysfunction in mouse pancreatic islets. Biochem J. 473:487–496. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Manukyan L, Ubhayasekera SJ, Bergquist J, Sargsyan E and Bergsten P: Palmitate-induced impairments of β-cell function are linked with generation of specific ceramide species via acylation of sphingosine. Endocrinology. 156:802–812. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Cnop M, Foufelle F and Velloso LA: Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med. 18:59–68. 2012.PubMed/NCBI View Article : Google Scholar | |
|
El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, Joly E, Dbaibo G, Rosenberg L and Prentki M: Saturated fatty acids synergize with elevated glucose to cause pancreatic beta-cell death. Endocrinology. 144:4154–4163. 2003.PubMed/NCBI View Article : Google Scholar | |
|
Wang XY, Zhu BR, Jia Q, Li YM, Wang T and Wang HY: Cinnamtannin D1 protects pancreatic β-cells from glucolipotoxicity-induced apoptosis by enhancement of autophagy in vitro and in vivo. J Agric Food Chem. 68:12617–12630. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Omar-Hmeadi M and Idevall-Hagren O: Insulin granule biogenesis and exocytosis. Cell Mol Life Sci. 78:1957–1970. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Back SH, Kang SW, Han J and Chung HT: Endoplasmic reticulum stress in the β-cell pathogenesis of type 2 diabetes. Exp Diabetes Res. 2012(618396)2012.PubMed/NCBI View Article : Google Scholar | |
|
Eizirik DL, Cardozo AK and Cnop M: The role for endoplasmic reticulum stress in diabetes mellitus. Endocr Rev. 29:42–61. 2008.PubMed/NCBI View Article : Google Scholar | |
|
Lee JH and Lee J: Endoplasmic reticulum (ER) stress and its role in pancreatic β-cell dysfunction and senescence in type 2 diabetes. Int J Mol Sci. 23(4843)2022.PubMed/NCBI View Article : Google Scholar | |
|
Hung CT, Tsai YW, Wu YS, Yeh CF and Yang KC: The novel role of ER protein TXNDC5 in the pathogenesis of organ fibrosis: mechanistic insights and therapeutic implications. J Biomed Sci. 29(63)2022.PubMed/NCBI View Article : Google Scholar | |
|
Wang X, Li H and Chang X: The role and mechanism of TXNDC5 in diseases. Eur J Med Res. 27(145)2022.PubMed/NCBI View Article : Google Scholar | |
|
Okumura M, Kadokura H and Inaba K: Structures and functions of protein disulfide isomerase family members involved in proteostasis in the endoplasmic reticulum. Free Radic Biol Med. 83:314–322. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Duivenvoorden WCM, Hopmans SN, Austin RC and Pinthus JH: Endoplasmic reticulum protein ERp46 in prostate adenocarcinoma. Oncol Lett. 13:3624–3630. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Bidooki SH, Navarro MA, Fernandes SCM and Osada J: Thioredoxin domain containing 5 (TXNDC5): Friend or Foe? Curr Issues Mol Biol. 46:3134–3163. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Rutter GA, Pullen TJ, Hodson DJ and Martinez-Sanchez A: Pancreatic β-cell identity, glucose sensing and the control of insulin secretion. Biochem J. 466:203–218. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Lampropoulou E, Lymperopoulou A and Charonis A: Reduced expression of ERp46 under diabetic conditions in β-cells and the effect of liraglutide. Metabolism. 65:7–15. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Cnop M, Abdulkarim B, Bottu G, Cunha DA, Cunha DA, Igoillo-Esteve M, Masini M, Turatsinze JV, Griebel T, Villate O, Santin I, et al: RNA sequencing identifies dysregulation of the human pancreatic islet transcriptome by the saturated fatty acid palmitate. Diabetes. 63:1978–1993. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, Wu J, Mbow CA, Jobava R, Haataja L, et al: Adaptation to chronic ER stress enforces pancreatic β-cell plasticity. Nat Commun. 13(4621)2022.PubMed/NCBI View Article : Google Scholar | |
|
Jiang H, Thapa P, Hao Y, Ding N, Alshahrani A and Wei Q: Protein disulfide isomerases function as the missing link between diabetes and cancer. Antioxid Redox Signal. 37 (16-18):1191–1205. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Taniguchi CM, Emanuelli B and Kahn CR: Critical nodes in signalling pathways: Insights into insulin action. Nat Rev Mol Cell Biol. 7:85–96. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Camaya I, Donnelly S and O'Brien B: Targeting the PI3K/Akt signaling pathway in pancreatic β-cells to enhance their survival and function: An emerging therapeutic strategy for type 1 diabetes. J Diabetes. 14:247–260. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Dalle S and Abderrahmani A: Receptors and signaling pathways controlling beta-cell function and survival as targets for anti-diabetic therapeutic strategies. Cells. 13(1244)2024.PubMed/NCBI View Article : Google Scholar | |
|
Back SH and Kaufman RJ: Endoplasmic reticulum stress and type 2 diabetes. Annu Rev Biochem. 81:767–793. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Hetz C and Papa FR: The unfolded protein response and cell fate control. Mol Cell. 69:169–181. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Sano R and Reed JC: ER stress-induced cell death mechanisms. Biochim Biophys Acta. 1833:3460–3470. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Hetz C, Zhang K and Kaufman RJ: Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. 21:421–438. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Ozcan U, Yilmaz E, Ozcan L, Furuhashi M, Vaillancourt E, Smith RO, Görgün CZ and Hotamisligil GS: Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science. 313:1137–1140. 2006.PubMed/NCBI View Article : Google Scholar | |
|
Gao Y, Ryu H, Lee H, Kim YJ, Lee JH and Lee J: ER stress and unfolded protein response (UPR) signaling modulate GLP-1 receptor signaling in the pancreatic islets. Mol Cells. 47(100004)2024.PubMed/NCBI View Article : Google Scholar | |
|
Xing D, Zhou Q, Wang Y and Xu J: Effects of tauroursodeoxycholic acid and 4-phenylbutyric acid on selenium distribution in mice model with type 1 diabetes. Biol Trace Elem Res. 201:1205–1213. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Yong J, Johnson JD, Arvan P, Han J and Kaufman RJ: Therapeutic opportunities for pancreatic β-cell ER stress in diabetes mellitus. Nat Rev Endocrinol. 17:455–467. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Ron D and Walter P: Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 8:519–529. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Celik C, Lee SYT, Yap WS and Thibault G: Endoplasmic reticulum stress and lipids in health and diseases. Prog Lipid Res. 89(101198)2023.PubMed/NCBI View Article : Google Scholar | |
|
Chen X and Cubillos-Ruiz JR: Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer. 21:71–88. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Ren J, Bi Y, Sowers JR, Hetz C and Zhang Y: Endoplasmic reticulum stress and unfolded protein response in cardiovascular diseases. Nat Rev Cardiol. 18:499–521. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Tarantino G and Citro V: Crosstalk between the spleen and other organs/systems: Downstream signaling events. Immuno. 4:479–501. 2024. | |
|
Zhang J, Deng Z, Jin L, Yang C, Liu J, Song H, Han W and Si Y: Spleen-derived anti-inflammatory cytokine IL-10 stimulated by adipose tissue-derived stem cells protects against type 2 diabetes. Stem Cells Dev. 26:1749–1758. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Gotoh K, Inoue M, Masaki T, Chiba S, Shimasaki T, Ando H, Fujiwara K, Katsuragi I, Kakuma T, Seike M, et al: A novel anti-inflammatory role for spleen-derived interleukin-10 in obesity-induced inflammation in white adipose tissue and liver. Diabetes. 61:1994–2003. 2012.PubMed/NCBI View Article : Google Scholar | |
|
Malik A, Bagchi AK, Jassal DS and Singal PK: Interleukin-10 mitigates doxorubicin-induced endoplasmic reticulum stress as well as cardiomyopathy. Biomedicines. 10(890)2022.PubMed/NCBI View Article : Google Scholar | |
|
Marafon BB, Pinto AP, de Sousa Neto IV, da Luz CM, Pauli JR, Cintra DE, Ropelle ER, Simabuco FM, Pereira de Moura L, de Freitas EC, et al: The role of interleukin-10 in mitigating endoplasmic reticulum stress in aged mice through exercise. Am J Physiol Endocrinol Metab. 327:E384–E395. 2024.PubMed/NCBI View Article : Google Scholar |