Pancreatic β-cells adapt to compensate for increased metabolic demand during obesity. Although the miRNA pathway has an essential role in β-cell expansion, whether it is involved in adaptive proliferation is largely unknown. First, we report that EGR2 binding to the miR-455 promoter induced miR-455 upregulation in the pancreatic islets of obesity mouse models. Then, in vitro gain- or loss-of-function studies showed that miR-455 overexpression facilitated β-cell proliferation. Knockdown of miR-455 in ob/ob mice via pancreatic intraductal infusion prevented compensatory β-cell expansion. Mechanistically, our results revealed that increased miR-455 expression inhibits the expression of its target cytoplasmic polyadenylation element binding protein 1 (CPEB1), an mRNA binding protein that plays an important role in regulating insulin resistance and cell proliferation. Decreased CPEB1 expression inhibits elongation of the poly(A) tail and the subsequent translation of Cdkn1b mRNA, reducing the CDKN1B expression level and finally promoting β-cell proliferation. Taken together, our results show that the miR-455/CPEB1/CDKN1B pathway contributes to adaptive proliferation of β-cells to meet metabolic demand during obesity.

Pancreatic β-cell function and mass are markedly adaptive to compensate for the changes in insulin requirements observed during obesity (1). Clinical results have shown that the β-cell mass in the pancreata of obese individuals without diabetes or with prediabetes is larger than that in lean individuals with normoglycemia (2). Evidence for this compensatory process has been consistently provided by obese rodent models and human pancreas necropsies (3,4). In obesity, β-cell mass increases by 30–40%, whereas insulin secretory output is augmented by 100% (5). Postmortem histology further reveals a 20–65% decrease in β-cell mass in islets from obese individuals with type 2 diabetes (T2D) compared with BMI-matched subjects without diabetes (6,7). This adaptive capacity of human islets to obesity has been confirmed in experimental murine models (8,9). Strategies aimed at improving β-cell function and mass plasticity could be of major interest for designing innovative therapeutics to prevent β-cell decline and restore β-cell functional adaptive ability in diabetes. However, the physiological mechanisms that promote adaptive pancreatic β-cell expansion are still not completely understood.

The adaptive capacity of β-cell mass and function depends on the activity of transcriptional and translational regulators, and miRNAs are extremely important in accomplishing this task (10,11). In pancreatic β-cells, miRNAs regulate insulin production by directly or indirectly affecting the expression of key transcription factors and contribute to fine-tuning of hormone release by modulating the levels of important components of the β-cell secretory machinery (1214). We and other researchers have reported changes in the expression of islet miRNAs in animal models of diabetes, with detrimental effects on the secretory activity and survival of β-cells (15,16). Moreover, obese mice lacking certain miRNAs failed to compensate for insulin resistance and developed a severe diabetic phenotype (3,12,17). These observations prompted us to investigate whether changes in miRNA expression contribute to compensatory β-cell mass expansion during obesity.

In this study, we found that miR-455 was upregulated during obesity to inhibit the expression of cytoplasmic polyadenylation element binding protein 1 (CPEB1) in pancreatic β-cells. Both in vivo and in vitro experimental results revealed that overexpression of (oe)-miR-455 promoted adaptive pancreatic β-cell proliferation and that knockdown of miR-455 prevented compensatory β-cell expansion. Moreover, we observed that the miR-455/CPEB1 pathway inhibits translation of cyclin-dependent kinase inhibitor 1b (Cdkn1b) mRNA and ultimately leads to lower levels of CDKN1B in β-cells, in turn significantly promoting β-cell adaptive proliferation. These observations show that miR-455 plays an integral role in the β-cell compensatory mechanism during obesity.

Animal Care

Eight-week-old C57BL/6J mice, ob/ob mice (4–12 weeks), and db/db mice (4–12 weeks) were obtained from GemPharmatech Co., Ltd. (Nanjing, China). All animals were on the C57BL/6 background unless otherwise stated. The db/db mice were on the BKS background. The care of all animals was within institutional animal care committee guidelines, and all procedures were approved by the animal ethics committee of China Pharmaceutical University (permit no. 2162326) and in accordance with international laws and policies (European Union Council Directive 86/609/EEC, 1987). Unless otherwise stated, C57BL/6J mice were fed a normal chow diet (10% calories from fat, D12450J; Research Diets) and provided water ad libitum. Diet-induced obesity was obtained by feeding a high-fat diet (HFD) (60% calories from fat, D12494; Research Diets) for at least 15 weeks. Male mice were used for all the indicated studies.

Isolation and Culture of Primary Islets

Six human islets were provided by Tianjin First Central Hospital. All human subjects provided informed consent, and all procedures using human islets were approved by the Research Ethics Committee of the Tianjin First Central Hospital (18). High-purity islets (>80%) were collected and cultured in CMRL-1066 medium (Corning, Manassas, VA) supplemented with 10% human serum albumin (Baxter, Vienna, Austria), 100 units/mL penicillin, and 100 μg/mL streptomycin at 37°C in 5% CO2. The islets from six human donors were mixed randomly, then mixed islets were used to perform each type of experiment and three technical replicates.

Mouse islets were isolated via collagenase digestion and enriched using a Histopaque (Sigma-Aldrich, St. Louis, MO) density gradient, as described previously (15,19). To assess the effects of pathophysiological concentrations of palmitate, glucose, and proinflammatory cytokines, islets were incubated in modified medium with 0.5% (w/v) BSA and various concentrations of glucose (low glucose 2.5 mmol/L, high glucose 25 mmol/L), palmitate (0.5 mmol/L), interleukin-1β (5 ng/mL), and tumor necrosis factor-α (30 ng/mL).

MIN6 Cell Culture

The mouse pancreatic β-cell line MIN6 (passages 15–20) was cultured in DMEM (Gibco, Burlington, MA) containing 15% FBS (Gibco), 100 IU/mL penicillin, 100 μg/mL streptomycin, and 50 μmol/L β-mercaptoethanol (Sigma-Aldrich) at 37°C in a humidified atmosphere containing 5% CO2.

Human EndoC-βH1 Cell Culture

Human EndoC-βH1 cells were obtained from Univercell Biosolutions (Toulouse, France). Human EndoC-βH1 cells were cultured in extracellular matrix/fibronectin-coated plates in low-glucose DMEM with supplements as previously described (20,21).

Insulin Secretion Assay

For the glucose-stimulated insulin secretion assay, MIN6 cells or human islets were preincubated overnight in Krebs-Ringer bicarbonate HEPES balanced buffer containing 0.2% BSA supplemented with 2.5 mmol/L glucose and incubated for 2 h with 2.5 or 16.7 mmol/L glucose. Immediately after incubation, an aliquot of the medium was removed for insulin analysis, and the cells were incubated in acid-ethanol for insulin content determination using a mouse insulin ELISA kit (ExCell Bio, Shanghai, China) according to the manufacturer’s instructions.

miRNA Sequencing and Analysis

Total RNA was isolated from islets of 8-week-old control mice and 8-week-old ob/ob mice using an RNeasy Plus Universal Mini Kit (QIAGEN), following the protocol for total RNA isolation. RNA integrity and concentration were assessed using an RNA Nano 6000 Assay Kit for the Bioanalyzer 2100 system (Agilent Technologies, Santa Clara, CA), according to the manufacturer’s instructions. To extract small RNA (18–30 nucleotides [nt]), total RNA was separated in 15% agarose gels. After ethanol precipitation and centrifugal enrichment of small RNA samples, a small RNA library was prepared using a Small RNA Sample Preparation Kit (RS-200-0048; Illumina), according to the manufacturer’s instructions. Briefly, a 3′ adaptor or 5′ adaptor was attached directionally. Each library was indexed with the Illumina adaptor (6-base barcode). The small RNA library was size fractionated in a 6% Tris base, boric acid, EDTA-urea polyacrylamide gel, and the 140- to 160-base pair (bp) fraction was excised from the gel. The library RNA concentration was measured using a Qubit RNA Assay Kit in Qubit 2.0 for preliminary quantification and then diluted to 1 ng/μL. Insert size was assessed using a 2100 Bioanalyzer System (Agilent Technologies), and after the insert size was confirmed to be consistent with expectations, the qualified insert size was accurately quantified using a TaqMan fluorescence probe and an Applied Biosystems StepOnePlus Real-Time PCR System (valid library concentration >2 nmol/L). Then, the qualified libraries were sequenced on an Illumina HiSeq 2500 platform. We calculated the fragments per kilobase per million mapped reads for mature miRNA from the mapped reads using a custom Python script. Differentially expressed miRNAs were analyzed using DESeq version 1.26.0 on the basis of the negative binomial distribution test (22), and the P value was corrected for false discovery rate analysis for multiple testing (23). Differentially expressed genes were considered significant with an adjusted P ≤ 0.05 and an absolute value of log2 (fold change) >1. The raw data are presented in Supplementary Table 1 and have been deposited in the National Center for Biotechnology Information Sequence Read Archive database (PRJNA731006).

Serum Samples of Subject Without Diabetes and With T2D

Serum and clinicopathological data were collected from Zhongda Hospital, which is affiliated with Southeast University. All human subjects provided informed consent, and all human studies were conducted according to the principles of the Declaration of Helsinki and approved by the ethics committees of Zhongda Hospital, Southeast University.

Electrophoretic Mobility Shift Assay Analysis

The probe, an ∼25-bp fragment, including the binding size, was biotin end labeled according to the instructions of the Pierce Biotin 3′ End DNA Labeling Kit (Thermo Fisher Scientific, Waltham, MA) and then annealed to double-stranded probe DNA. Egr2-DNA complexes were generated according to the instructions of a LightShift Chemiluminescent EMSA Kit (Thermo Fisher Scientific). Probe sequences are listed in Supplementary Table 2.

Chromatin Immunoprecipitation Experiment

MIN6 cells and human islets were fixed with 37% formaldehyde for 10 min, followed by 30 rounds of sonication (each for 3 s) to fragment the chromatin. Chromatin was incubated with an anti–early growth response 1 (EGR2) antibody at 4°C overnight and then immunoprecipitated with Proteinase K (EMD Millipore, Billerica, MA). Purified DNA was amplified via quantitative RT-PCR (qRT-PCR) using PCR and primer pairs that spanned the predicted Egr2 binding sites. Primer sequences are listed in Supplementary Table 2.

Plasmid Construction

The coding sequences for Egr2 (NM_001373983.1), Cpeb1 (NM_001252525.1), and Cdkn1b (NM_009875.4) were amplified via PCR from full-length cDNA of MIN6 cells and then cloned into a pcDNA 3.1 vector. The primer sequences for PCR are listed in Supplementary Table 2.

The single-guide RNAs (sgRNAs) for the miR-455 promoter were constructed in a lentiCRISPRv2 puro vector according to the Zhang libraries (24). The lentiCRISPRv2 puro vector was digested with BsmBI. The sgRNA seed sequences are listed in Supplementary Table 3.

Luciferase Assays

MIN6 cells (2 × 105 cells/well) were transfected with 0.4 μg miR-455 promoter, 0.4 μg Egr2, and 0.1 μg constitutive Renilla expression plasmid, and luciferase activities were measured using a dual-luciferase reporter assay system (Vazyme, Nanjing, China) after transfection for 24 h. The complete 3′-untranslated region (UTR) of murine Cpeb1 containing either the wild-type (WT) or the mutated (MUT) miR-455 binding site was cloned behind the stop codon of the firefly luciferase open reading frame using specific primers. MIN6 cells were transfected with pmirPGLO reporter (100 ng) along with miR-455 or miR-455 inhibitor using Lipofectamine 2000 transfection reagent (Invitrogen). At 24 h posttransfection, dual luciferase reporter assays were performed using a luciferase assay system (Vazyme). The WT and MUT site primers are listed in Supplementary Table 2.

Flow Cytometric Analysis of Proliferation

Insulin and KI-67 double-positive cells were detected following a standard intracellular staining procedure using Cytofix/Cytoperm solution (BD Biosciences). Insulin-allophycocyanin (APC) and KI-67-FITC antibodies were used for staining, and isotype antibodies were used as a negative control (NC). After double staining with insulin-APC and KI-67-FITC, the cells were analyzed with a flow cytometer (FACScan; BD Biosciences) equipped with FlowJo version 10 software (BD Biosciences).

β-Cell Purification Via Flow Cytometric Analysis

Purified islets were incubated with 2 mg/mL collagenase II (Sigma-Aldrich) for 10 min, and insulin-positive cells were detected by following a standard intracellular staining procedure using Cytofix/Cytoperm solution. After staining with insulin-APC, the cells were analyzed via flow cytometry (BD FACSAria II SORP). Purified β-cell and non–β-cell fractions were collected for RNA extraction and qRT-PCR analysis.

Pancreatic Intraductal Viral Infusion in Mice

miR-455, miR-455 inhibitor, sh-Cpeb1, and sh-Cdkn1b were inserted into a lentivirus-mouse insulin 2 promoter (MIP)-PGLV3/H1/puro vector (len-miR-455, len-anti-miR-455, len-shCpeb1, len-shCdkn1b). The indicated lentivirus (1 × 109) was dissolved in 0.15 mL normal saline and infused into the pancreatic duct at 6 μL/min for 25 min using an R462 perfusion pump (RWD Life Sciences, Shengzhen, China), according to the procedure described by Xiao et al. (25). At 72 h after injection, islets were lysed to extract total RNA or protein to measure the overexpression efficacy. At 1 week postinjection, the mice were fed an HFD (60% energy from fat, D12494; Research Diets) for 20 weeks.

Mouse Metabolic Assays

Mouse fasting blood glucose levels and fasting serum insulin levels were examined using a glucometer (OMRON, Kyoto, Japan) and an ELISA (ExCell Bio) after 12 h of fasting treatment. To perform the glucose tolerance tests, 1.5 g/kg glucose (Sigma-Aldrich) was injected i.p. into mice, whereas 0.75 units/kg insulin (Novolin R; Novo Nordisk, Bagsværd, Denmark) was injected i.p. into mice for insulin tolerance tests.

RNA Extraction and qRT-PCR Analysis

Total RNA was extracted using TRIzol (Invitrogen), as previously described (17). RT reactions were performed using a PrimeScript RT Reagent Kit (Takara Bio, Tokyo, Japan), and diluted cDNA was used for qRT-PCR analysis using a SYBR Premix Ex Taq II Kit (Takara Bio) with the appropriate primers listed in Supplementary Table 4. The relative expression of genes was determined using a comparative method (2−ΔCT). miRNA and mRNA levels were normalized to the expression of small RNAs (sno234 and U6) or mRNA (Gapdh, Hprt, and Ppia), respectively. For miR-455 and U6, TaqMan probes (Ambion) were used to confirm our results.

PCR Poly(A) Tail Length Assay

Total cellular RNA was reversed transcribed using MultiScribe RT (Life Technologies) and an oligo (dT) anchor primer (5′-GCGAGCTCCGCGGCCGCGTTTTTTTTTTTTTTT-3′), and subsequent PCR was conducted with an anchor primer (5′-AAAAACGCGGGCCGCGGAGCTCGC-3′) and a specific primer for Cdkn1b (5′-GCCAATTATTGTTACACATT-3′) located near the 3′ end of the Cdkn1b 3′-UTR.

RNA Immunoprecipitation

RNA immunoprecipitation (RIP) was performed using an EZMagna RIP Kit (EMD Millipore), according to the manufacturer’s protocol. Cells were lysed in complete RIP lysis buffer, and then 100 μL of whole-cell extract was incubated with RIP buffer containing magnetic beads conjugated with anti-Ago2 (Cell Signaling Technology) or anti-CPEB1 (Abcam) antibody or NC normal mouse IgG (Abcam). Furthermore, purified RNA was subjected to qRT-PCR analysis to demonstrate the presence of the binding targets using the respective primers. The primer sequences are listed in Supplementary Table 4.

Immunohistochemistry and Immunofluorescence

Pancreata were fixed in 4% paraformaldehyde and embedded in paraffin, and the antigen in the cut sections was retrieved by boiling in 10 mmol/L Tris/EDTA (pH 9.0). Sections were permeabilized and blocked in PBS buffer containing 0.3% Triton X-100, 1% BSA, and 5% goat serum. Primary antibody (anti-KI-67, anti-insulin, antiglucagon) binding was performed overnight at 4°C, and incubation with secondary antibody was performed at room temperature for 1 h. The slides were analyzed using confocal laser scanning microscopy (CLSM) (LSM 700; Carl Zeiss) at ×20 or ×40 magnification. The antibodies are listed in Supplementary Table 5.

Fluorescence In Situ Hybridization

A Cy3-labeled miR-455 probe was designed and synthesized by GenePharma (Shanghai, China). For the fluorescence in situ hybridization assay, pancreata were fixed in 4% formaldehyde, permeabilized with 0.3% Triton X-100 for 15 min, and washed with PBS three times and once in 2× saline-sodium citrate buffer. Hybridization was carried out at 37°C for 16 h using DNA probe sets, followed by incubation with anti-insulin antibody overnight at 4°C and incubation with secondary antibody at room temperature for 1 h. Images were obtained with a CLSM and processed using ZEN imaging software.

Western Blot Analysis

Mouse islets (n = 200 islets/group) and MIN6 cells were lysed with radioimmunoprecipitation assay lysis buffer (Beyotime) containing 1% phenylmethylsulfonyl fluoride (Sigma-Aldrich). Then, Western blot analyses were conducted according to standard procedures using specific antibodies. The antibodies are listed in Supplementary Table 5.

Insulin/KI-67 Double Staining

MIN6 cells and primary islets were fixed in 4% paraformaldehyde and permeabilized and blocked in PBS buffer containing 0.3% Triton X-100 and 1% BSA. Primary insulin and KI-67 antibody binding was performed overnight at 4°C, and then incubation with secondary antibody was performed at room temperature for 1 h. Finally, the nuclei were stained with DAPI. Images were obtained using a CLSM at ×40 or ×20 magnification.

β-Cell Mass

Pancreatic sections were stained with anti-insulin antibody and DAPI to determine β-cell mass. The pancreatic sections were scanned entirely using the 10× objective of a Zeiss LSM 700 microscope. β-Cell mass was calculated by multiplying the area of insulin-positive cells/pancreas area using ImageJ software (26).

Cell Counting Kit-8 Assay

MIN6 cells were seeded into 96-well plates (4 × 104 cells/well) in 100 μL culture medium. A Cell Counting Kit-8 assay (Vazyme) was performed according to the manufacturer’s instructions.

Statistical Analysis

All data represent at least three independent experiments and are shown as the mean ± SD. Comparisons between two groups were performed using Student t test and among multiple groups using ANOVA. Dunn multiple comparisons for one-way ANOVA and Fisher least significant difference for two-way ANOVA were used. P < 0.05 was considered significant. GraphPad Prism 7 software (GraphPad Software, San Diego, CA) was used for all calculations.

Data and Resource Availability

The data sets generated and/or analyzed during the current study are available from the corresponding authors upon reasonable request. The RNA sequencing raw data that support the findings of this study have been deposited in the National Center for Biotechnology Information Sequence Read Archive database (PRJNA731006; https://submit.ncbi.nlm.nih.gov/subs/sra/SUB9682154/overview).

miR-455 Is Elevated in the Islets of Obese Mouse Models

To identify miRNAs potentially involved in β-cell mass expansion during obesity, we performed small RNA sequencing on total RNA from 8-week-old ob/ob mice, which are capable of long-term compensatory insulin hypersecretion (27). The characteristics of the animals used in this study are presented in Supplementary Fig. 1A and B. Using an absolute fold change of at least 1.0 and P < 0.05, we observed that the expression of 39 miRNAs was significantly upregulated and that the expression of 46 miRNAs was downregulated in ob/ob mice compared with WT mice. Among the increased miRNAs identified, the expression of miR-6932-3p (miR-6932) and miR-455-5p (miR-455) was the most significantly increased (Fig. 1A and Supplementary Table 1). miR-6932 expression was significantly lower than miR-455 expression in human islets, mouse islets, and MIN6 cells (Supplementary Fig. 1C). Therefore, we chose miR-455 for further analysis. Then, we measured the expression of miR-455 in the islets of ob/ob mice aged 4–12 weeks and observed an increase in miR-455 expression at 6 weeks of age with the onset of insulin resistance (Fig. 1B). Similarly, the pri-miR-455 transcript was also upregulated in the islets of ob/ob mice (Fig. 1C), indicating that miR-455 is regulated at the transcriptional level. We observed a similar increase in the expression of mature miR-455 in the islets of mice treated with a 15-week HFD, where miR-455 expression was significantly decreased, accompanied by an increase in glucose (Fig. 1D). We also found that miR-455 expression was dramatically increased in the islets of young db/db mice (Fig. 1E), all of which showed that this observation is not limited to one mouse model of obesity and insulin resistance. The expression of miR-455 was only significantly increased in islets, white adipose tissue (WAT), brown adipose tissue (BAT), and muscle in ob/ob and HFD mice compared with their respective controls (Fig. 1F). Interestingly, we also found that miR-455 levels were significantly increased in donors with T2D compared with donors without diabetes (Fig. 1G), and serum miR-455 expression levels were significantly correlated with BMI and HbA1c in these subjects (Fig. 1H and I). Then, we found that miR-455 expression was approximately fourfold higher than that in exocrine glands, indicating that islets represent the main source of miR-455 in the pancreas (Fig. 1J and K). Moreover, we purified β-cells from islets via flow cytometric analysis, and the results revealed that miR-455 was enriched in purified β-cells (Fig. 1L). Overall, miR-455 expression in islets is increased in both dietary and genetic mouse models of obesity.

Figure 1

miR-455 is elevated in the islets of obese mouse models. A: Comparison of small RNA sequencing analysis of total RNA from islets of 8-week-old ob/ob mice and WT littermates (n = 6 mice/group). B and C: qRT-PCR was used to detect the expression levels of miR-455 (B) and pri-miR-455 (C) in the islets of 4–12-week-old ob/ob mice and control mice (n = 3 mice/group). D: miR-455 levels in the islets of C57BL/6J mice treated with a 15-week HFD. The glucose concentrations indicate blood glucose levels in the mice (normal chow diet [NCD], n = 3 mice/group). E: miR-455 levels in the islets of 4–12-week-old db/db mice (n = 3 mice/group). F: The expression levels of miR-455 in different tissues from ob/ob mice and HFD mice compared with WT mice and NCD mice, respectively (n = 3 mice/group). G: The expression levels of miR-455 in the serum extracted from individuals without diabetes (n = 21) and with T2D (n = 76) using Ce-miR-39-1 as positive control. H and I: Correlation between miR-455 levels and BMI or HbA1c. Pearson correlation coefficients (R) are shown. J: miR-455 expression levels in the pancreas, islets, and exocrine glands of C57BL/6J mice (n = 3 mice/group). K: Immunofluorescence was performed to determine the miR-455 expression levels in the pancreas of C57BL/6J mice. Magnification, ×20; scale bar, 20 μm. L: The levels of miR-455 in the islet, purified β-cell, and non–β-cell fractions (n = 5 mice/group). Data are mean ± SD. **P < 0.01, ***P < 0.001.

Figure 1

miR-455 is elevated in the islets of obese mouse models. A: Comparison of small RNA sequencing analysis of total RNA from islets of 8-week-old ob/ob mice and WT littermates (n = 6 mice/group). B and C: qRT-PCR was used to detect the expression levels of miR-455 (B) and pri-miR-455 (C) in the islets of 4–12-week-old ob/ob mice and control mice (n = 3 mice/group). D: miR-455 levels in the islets of C57BL/6J mice treated with a 15-week HFD. The glucose concentrations indicate blood glucose levels in the mice (normal chow diet [NCD], n = 3 mice/group). E: miR-455 levels in the islets of 4–12-week-old db/db mice (n = 3 mice/group). F: The expression levels of miR-455 in different tissues from ob/ob mice and HFD mice compared with WT mice and NCD mice, respectively (n = 3 mice/group). G: The expression levels of miR-455 in the serum extracted from individuals without diabetes (n = 21) and with T2D (n = 76) using Ce-miR-39-1 as positive control. H and I: Correlation between miR-455 levels and BMI or HbA1c. Pearson correlation coefficients (R) are shown. J: miR-455 expression levels in the pancreas, islets, and exocrine glands of C57BL/6J mice (n = 3 mice/group). K: Immunofluorescence was performed to determine the miR-455 expression levels in the pancreas of C57BL/6J mice. Magnification, ×20; scale bar, 20 μm. L: The levels of miR-455 in the islet, purified β-cell, and non–β-cell fractions (n = 5 mice/group). Data are mean ± SD. **P < 0.01, ***P < 0.001.

Close modal

Silencing of Egr2 in Pancreatic β-Cells Suppresses miR-455 Expression

To determine the potential reason for the changes in miR-455 expression detected in the islets of obese mice, MIN6 cells, EndoC-βH1 cells, normal mouse islets, and human islets were exposed to pathophysiological concentrations of palmitate, glucose, and proinflammatory cytokines. The expression of miR-455 was increased in the presence of palmitate (0.5 mmol/L) (Fig. 2A–D) and a high glucose level (25 mmol/L) (Fig. 2E–H), but these changes were not observed in MIN6 cells or mouse islets incubated with proinflammatory cytokines (Supplementary Fig. 2AC).

Figure 2

Silencing of Egr2 in pancreatic β-cells represses miR-455 expression. AH: qRT-PCR was performed to examine the miR-455 expression levels in MIN6 cells, EndoC-βH1 cells, mouse primary islets and human islets incubated with 0.5 mmol/L palmitate (AD) or with 25 mmol/L glucose (EH). IL: The protein and mRNA levels of EGR2 in the islets of HFD (I) (n = 5 mice/group) and ob/ob (J) (n = 5 mice/group) mice as well as in the EndoC-βH1 cells incubated with 0.5 mmol/L palmitate (K) or 25 mmol/L glucose (L). MO: Enrichment of Egr2 on the miR-455 promoter relative to IgG in the islets of ob/ob mice (M), EndoC-βH1 cells (N), and human islets (O) transfected with oe-Egr2, pcDNA 3.1 vector, si-Egr2, or si-NC detected by chromatin immunoprecipitation-qPCR assays. PR: Direct binding of Egr2 to the miR-455 promoter in mouse islets (P) (n = 5 mice/group), EndoC-βH1 cells (Q), and human islets (R) determined by EMSA. C1 and C2 represent nuclear protein-miR-455 probe complexes and nuclear protein-miR-455 probe-anti-EGR2 complexes, respectively. S and T: The miR-455 expression levels in MIN6 cells (S) and human islets (T) incubated with 0.5 mmol/L palmitate and cotransfected with si-Egr2. Data are mean ± SD. **P < 0.01, ***P < 0.001. H-Islet, human islets; M-Islet, mouse islets; NS, not significant.

Figure 2

Silencing of Egr2 in pancreatic β-cells represses miR-455 expression. AH: qRT-PCR was performed to examine the miR-455 expression levels in MIN6 cells, EndoC-βH1 cells, mouse primary islets and human islets incubated with 0.5 mmol/L palmitate (AD) or with 25 mmol/L glucose (EH). IL: The protein and mRNA levels of EGR2 in the islets of HFD (I) (n = 5 mice/group) and ob/ob (J) (n = 5 mice/group) mice as well as in the EndoC-βH1 cells incubated with 0.5 mmol/L palmitate (K) or 25 mmol/L glucose (L). MO: Enrichment of Egr2 on the miR-455 promoter relative to IgG in the islets of ob/ob mice (M), EndoC-βH1 cells (N), and human islets (O) transfected with oe-Egr2, pcDNA 3.1 vector, si-Egr2, or si-NC detected by chromatin immunoprecipitation-qPCR assays. PR: Direct binding of Egr2 to the miR-455 promoter in mouse islets (P) (n = 5 mice/group), EndoC-βH1 cells (Q), and human islets (R) determined by EMSA. C1 and C2 represent nuclear protein-miR-455 probe complexes and nuclear protein-miR-455 probe-anti-EGR2 complexes, respectively. S and T: The miR-455 expression levels in MIN6 cells (S) and human islets (T) incubated with 0.5 mmol/L palmitate and cotransfected with si-Egr2. Data are mean ± SD. **P < 0.01, ***P < 0.001. H-Islet, human islets; M-Islet, mouse islets; NS, not significant.

Close modal

Next, we explored the potential molecular mechanism of miR-455 upregulation in the islets of obese mice. We predicted an miR-455 promoter region 2 kb upstream of the mouse and human miR-455 sequence and constructed three sgRNAs corresponding to this promoter in a lentiCRISPRv2 puro vector. Supplementary Fig. 2D and E show that miR-455 levels were decreased in MIN6 cells and human islets transfected with sgRNAs. These results revealed that the predicted promoter region can modify miR-455 expression. Then, MIN6 cells were exposed to 0.5 mmol/L palmitate for 48 h, and qRT-PCR was performed to confirm the expression levels of high-score transcription factors (score >15) of the miR-455 promoter, which were predicted by JASPAR. Among these candidates, Egr2, which has two potential binding sites in the mus-miR-455 promoter (Supplementary Fig. 2F), was mostly upregulated (Supplementary Fig. 2G). Interestingly, Egr2 also has one potential binding site in the hsa-miR-455 promoter (Supplementary Fig. 2F). Moreover, the mRNA and protein levels of EGR2 were upregulated in obese mouse islets (Fig. 2G and I) and MIN6 cells in response to palmitate treatment (Supplementary Fig. 2H) or high-glucose treatment (Supplementary Fig. 2I). The expression levels of EGR2 were also increased in human islets (Supplementary Fig. 2J and K) and EndoC-βH1 cells (Fig. 2K and L) exposed to palmitate and high glucose.

Egr2 is known as an important regulator of insulin resistance (28) and can be upregulated by palmitate in MIN6 cells (29). We next focused on studying whether obesity facilitates miR-455 upregulation by increasing Egr2 expression. As expected, in dual luciferase assays, MIN6 cells transfected with Egr2 overexpression plasmid exhibited a significantly higher Egr2 binding ability to the miR-455 promoter compared with the control, and after mutation of the R1 binding region, the binding ability was significantly decreased (Supplementary Fig. 2L). Moreover, Egr2 in the miR-455 promoter region was increased in the islets of obese mice (Fig. 2M and Supplementary Fig. 2M). A similar trend was obtained in MIN6 cells (Supplementary Fig. 2N and O), in EndoC-βH1 cells (Fig. 2N) and human islets (Fig. 2O) transfected with oe-Egr2. EMSA results revealed that the signal from the probe-protein-anti-EGR2 complex could be detected using an miR-455 probe in mouse islets (Fig. 2P), EndoC-βH1 cells (Fig. 2Q), human islets (Fig. 2R), and MIN6 cells (Supplementary Fig. 2P). Further research showed that palmitate-induced miR-455 upregulation in MIN6 cells and human islets was partially reversed by knockdown of Egr2 (Fig. 2S and T). These results indicate that the increased miR-455 expression in the islets of obese mice is mediated by upregulation of Egr2.

miR-455 Regulates β-Cell Proliferation

To explore the potential miR-455 function in β-cells, MIN6 cells were transfected with miR-455 mimic (miR-455) or miR-455 inhibitor (anti-miR-455). The knockdown and overexpression efficiencies were ∼80% and 220-fold, respectively (Supplementary Fig. 3A). Cell Counting Kit-8 assay results showed that cell proliferation was reduced by miR-455 silencing (Supplementary Fig. 3B). We observed that the upregulation of miR-455 in MIN6 cells resulted in an increase in the number of insulin-positive/Ki-67+ cells (Fig. 3A). Similar results were obtained in MIN6 cells (Fig. 3B), EndoC-βH1 cells (Fig. 3C), human islets (Fig. 3D), and mouse islets (Supplementary Fig. 3C). Next, we assessed whether miR-455 regulates MIN6 cell proliferation by evaluating cell-cycle status. Flow cytometry results indicated that miR-455 reduced the proportion of MIN6 cells in the G1 phase and increased the proportion in the S phase (Fig. 3E). Since cell-cycle progression is tightly regulated by cyclin-related proteins, we next tested the expression levels of cyclin-related proteins in MIN6 cells transfected with miR-455 or anti-miR-455. As shown in Supplementary Fig. 3D, miR-455 significantly decreased the expression level of CDKN1B, which is an inhibitor of the G1-S transition. The CDKN1B protein level was also decreased in MIN6 cells transfected with miR-455 (Fig. 3F). However, miR-455 did not significantly affect insulin synthesis, insulin content, or insulin secretion (Supplementary Fig. 3EG) in MIN6 cells. Similar results were observed in human islets (Supplementary Fig. 3HJ).

Figure 3

miR-455 regulates β-cell proliferation. A and B: MIN6 cells were transfected with miR-455 mimic or miR-455 inhibitor (anti-miR-455) for 48 h. Then, insulin-positive/KI-67+ cells were counted via flow cytometry (A) and immunofluorescence (B). Magnification, ×20; scale bar, 20 μm. C and D: The insulin-positive/KI-67+ cells in EndoC-βH1 cells (C) and human islets (D) transfected with miR-455 or anti-miR-455 were counted via immunofluorescence. Magnification, ×20; scale bar, 20 μm. E: The percentage of cells in G1, S, or G2 phase was determined by flow cytometry. F: The CDKN1B protein level. G: The len-miR-455 or len-anti-miR-455 was injected via pancreatic ductal infusion for 72 h, and the miR-455 expression levels in the islets were detected by qRT-PCR (n = 3 mice/group). H: Intraperitoneal glucose tolerance test (IPGTT) (1.5 g/kg) in overnight-fasted len-miR-455 mice, len-anti-miR-455 mice, and NC mice (n = 9 mice/group). I: In vivo insulin excursions of overnight-fasted len-miR-455 mice, len-antimiR-455 mice, and NC mice after IPGTT exposure (n = 9 mice/group). J: KI-67+ β-cells in the islets of len-miR-455 mice and len-anti-miR-455 mice. Magnification, ×20; scale bar, 20 μm. K and L: CDKN1B protein levels in the islets of len-miR-455 mice and len-anti-miR-455 mice. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001. AUC, area under the curve; H-Islet, human islets.

Figure 3

miR-455 regulates β-cell proliferation. A and B: MIN6 cells were transfected with miR-455 mimic or miR-455 inhibitor (anti-miR-455) for 48 h. Then, insulin-positive/KI-67+ cells were counted via flow cytometry (A) and immunofluorescence (B). Magnification, ×20; scale bar, 20 μm. C and D: The insulin-positive/KI-67+ cells in EndoC-βH1 cells (C) and human islets (D) transfected with miR-455 or anti-miR-455 were counted via immunofluorescence. Magnification, ×20; scale bar, 20 μm. E: The percentage of cells in G1, S, or G2 phase was determined by flow cytometry. F: The CDKN1B protein level. G: The len-miR-455 or len-anti-miR-455 was injected via pancreatic ductal infusion for 72 h, and the miR-455 expression levels in the islets were detected by qRT-PCR (n = 3 mice/group). H: Intraperitoneal glucose tolerance test (IPGTT) (1.5 g/kg) in overnight-fasted len-miR-455 mice, len-anti-miR-455 mice, and NC mice (n = 9 mice/group). I: In vivo insulin excursions of overnight-fasted len-miR-455 mice, len-antimiR-455 mice, and NC mice after IPGTT exposure (n = 9 mice/group). J: KI-67+ β-cells in the islets of len-miR-455 mice and len-anti-miR-455 mice. Magnification, ×20; scale bar, 20 μm. K and L: CDKN1B protein levels in the islets of len-miR-455 mice and len-anti-miR-455 mice. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001. AUC, area under the curve; H-Islet, human islets.

Close modal

In view of these findings, to verify whether ectopic expression of miR-455 also affects β-cell proliferation in vivo, 1 × 109len-miR-455 cells or 1 × 109len-anti-miR-455 cells were injected into 8-week-old male C57BL/6J mice through pancreatic intraductal infusion. We observed an ∼65-fold miR-455 upregulation in the islets that received len-miR-455 compared with those that received lentivirus-MIP-miR-NC (len-miR-NC) and an ∼70% decrease in miR-455 expression in the islets of len-anti-miR-455 mice compared with len-anti-NC mice (Fig. 3G), whereas the expression level of miR-455 was not significantly changed in other organs (Supplementary Fig. 3K). The len-miR-455 mice exhibited slightly improved glucose tolerance in the intraperitoneal glucose tolerance test (Fig. 3H). Similarly, the insulin levels in len-miR-455 mice improved after glucose injection compared with those in control mice (Fig. 3I), while miR-455 had no effect on glucagon in vivo (Supplementary Fig. 3L). The len-miR-455 mice showed a significant increase in both β-cell proliferation (Fig. 3J) and β-cell mass (Supplementary Fig. 3M). Moreover, the CDKN1B mRNA and protein levels were suppressed in the islets of len-miR-455 mice and vice versa (Supplementary Fig. 3N and Fig. 3K and L). Furthermore, no change was detected in TUNEL+ β-cells after ectopic expression of miR-455 (Supplementary Fig. 3O), indicating that cell death was not a primary mechanism underlying the effect on β-cell mass.

Loss of miR-455 Expression in Obese Mice Inhibits β-Cell Proliferation

To address whether miR-455 mediates the compensatory expansion of β-cells during insulin resistance, 1 × 109len-anti-miR-455 cells were injected into 8-week-old male C57BL/6 mice via pancreatic ductal infusion, and then, the mice were fed an HFD for 20 weeks (Fig. 4A). As shown in Fig. 4B, miR-455 was downregulated to 70% in the islets of len-anti-miR-455 mice compared with those receiving len-anti-NC even 20 weeks after injection. The len-anti-miR-455 treatment had no effect on cumulative energy intake (Supplementary Fig. 4A), body weight (Supplementary Fig. 4B), or body fat content (Supplementary Fig. 4C). However, inhibition of miR-455 slowly increased glycemia in random-fed mice after 20 weeks of HFD treatment (Supplementary Fig. 4D). The obesity-associated rise in serum insulin concentrations was slightly lower in HFD-fed animals treated with len-anti-miR-455 (Fig. 4C), and HOMA of insulin resistance indices in mice with miR-455 repression were increased (Fig. 4D). In accordance with this, glucose tolerance tests revealed impairment of glucose tolerance upon miR-455 knockdown (Fig. 4E), and insulin sensitivity was damaged upon downregulation of miR-455 (Fig. 4F). Counts of KI-67+ β-cells were lower in len-anti-miR-455 mice than in control animals (Fig. 4G). Moreover, we found that miR-455 knockdown markedly decreased β-cell mass compared with that in len-anti-NC mice (Supplementary Fig. 4E). These findings indicate that the miR-455 inhibitor resulted in damage to β-cell mass in diet-induced obese mice.

Figure 4

Loss of miR-455 expression in obese mice inhibits β-cell proliferation. A: Flowchart of the in vivo experiments designed for detection of β-cell function after pancreatic ductal infusion. Eight-week-old male len-anti-miR-455 mice and control mice (lentivirus-MIP-LV3/H1 vector) were exposed to an HFD for 20 weeks. B: The miR-455 expression levels were examined via qRT-PCR. C and D: Fasting insulin levels in HFD-fed mice were measured via ELISA (C) (n = 9 mice/group) and HOMA of insulin resistance (HOMA-IR) index was measured (D) (n = 9 mice/group). The HOMA-IR index was calculated using the equation (fasting blood glucose [mmol/L] × fasting serum insulin [mIU/L]) / 22.5. E and F: Intraperitoneal glucose tolerance tests (IPGTTs) (1.5 g/kg) (E) and intraperitoneal insulin tolerance tests (IPITTs) (0.75 units/kg) (F) were performed in len-anti-miR-455 mice and NC mice at the 8th or 10th week of HFD administration, respectively. The corresponding area under the curve (AUC) of the blood glucose level was calculated (n = 9 mice/group). G: KI-67+ β-cells were analyzed by immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. H: Random blood glucose level in 8-week-old ob/ob mice treated with len-anti-miR-455 (n = 7 mice/group). I: Plasma insulin concentrations in 8-week-old ob/ob mice treated with len-anti-miR-455 (n = 7 mice/group). J: The number of KI-67+ β-cells in 8-week-old len-anti-miR-455/ob mice (n = 3 mice/group). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001.

Figure 4

Loss of miR-455 expression in obese mice inhibits β-cell proliferation. A: Flowchart of the in vivo experiments designed for detection of β-cell function after pancreatic ductal infusion. Eight-week-old male len-anti-miR-455 mice and control mice (lentivirus-MIP-LV3/H1 vector) were exposed to an HFD for 20 weeks. B: The miR-455 expression levels were examined via qRT-PCR. C and D: Fasting insulin levels in HFD-fed mice were measured via ELISA (C) (n = 9 mice/group) and HOMA of insulin resistance (HOMA-IR) index was measured (D) (n = 9 mice/group). The HOMA-IR index was calculated using the equation (fasting blood glucose [mmol/L] × fasting serum insulin [mIU/L]) / 22.5. E and F: Intraperitoneal glucose tolerance tests (IPGTTs) (1.5 g/kg) (E) and intraperitoneal insulin tolerance tests (IPITTs) (0.75 units/kg) (F) were performed in len-anti-miR-455 mice and NC mice at the 8th or 10th week of HFD administration, respectively. The corresponding area under the curve (AUC) of the blood glucose level was calculated (n = 9 mice/group). G: KI-67+ β-cells were analyzed by immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. H: Random blood glucose level in 8-week-old ob/ob mice treated with len-anti-miR-455 (n = 7 mice/group). I: Plasma insulin concentrations in 8-week-old ob/ob mice treated with len-anti-miR-455 (n = 7 mice/group). J: The number of KI-67+ β-cells in 8-week-old len-anti-miR-455/ob mice (n = 3 mice/group). Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001.

Close modal

Furthermore, high expression of miR-455 in the islets of ob/ob mice was silenced by len-anti-miR-455 through pancreatic intraductal infusion (Supplementary Fig. 4F). Compared with len-anti-NC/ob mice, len-anti-miR-455/ob mice exhibited severe hyperglycemia and reduced systemic insulin levels (Fig. 4H and I) due to loss of β-cell mass and compensation (Fig. 4J and Supplementary Fig. 4G). Collectively, these data show that suppression of miR-455 expression in obesity model mice attenuated adaptive β-cell proliferation.

miR-455 Affects β-Cell Proliferation by Targeting Cpeb1

Most miRNAs function by suppressing their target gene expression, and we found an inverse expression pattern between miR-455 and Cdkn1b, which prompted us to verify whether Cdkn1b is a target gene of miR-455. Unfortunately, using a double luciferase reporter, we found no targeting relationship between miR-455 and Cdkn1b (Supplementary Fig. 5A).

To identify the potential miRNA targets of miR-455, in silico analysis was performed using TargetScan, miRDB, miRWalk, and starBase, which jointly predicted that two genes (Cpeb1 and Usp9x) may act as biological targets of miR-455 (Fig. 5A). Next, we verified the targeting relationship between the two genes and miR-455. Cpeb1 harbored an miR-455 binding site, which was conserved in humans, mice, and rats (Fig. 5B). We cloned a portion of the mouse 3′-UTR of Cpeb1 (1,316 nt) into a luciferase reporter construct and observed decreased activity in the presence of an miR-455 mimic (Fig. 5C). In addition, mutation of 8 nt in the binding site within the mouse UTR (at positions 1,289–1,296) from GGCACAUA to AATGTGCG abolished the inhibitory effect of miR-455 (Fig. 5C). Moreover, we conducted anti-Ago2 RIP in MIN6 cells transiently overexpressing miR-455. Endogenous Cpeb1 pulldown by Ago2 was specifically enriched in miR-455–transfected cells (Fig. 5D) and vice versa (Supplementary Fig. 5B). oe-miR-455 decreased CPEB1 expression in the MIN6 cells (Supplementary Fig. 5C). The mRNA and protein levels of CPEB1 were also significantly decreased in the islets of len-miR-455 mice but increased in the islets of len-anti-miR-455 mice (Fig. 5E and F and Supplementary Fig. 5D). Finally, silencing the expression of miR-455 in the pancreas of ob/ob mice increased CPEB1 expression (Fig. 5G and Supplementary Fig. 5E). Unfortunately, using the same technical means, we did not find a targeted relationship between miR-455 and Usp9x (Supplementary Fig. 5F).

Figure 5

miR-455 affects β-cell proliferation by targeting Cpeb1. A: Four independent miRNA target prediction algorithms were used to predict the target genes of miR-455. B: Graphic representation of the conserved miR-455 binding motif in the Cpeb1 3′-UTR of three mammalian species. Binding of the WT (top) and mutated (bottom) murine Cpeb1-3′-UTR to the miR-455 seed sequence was assessed in reporter gene experiments. C: Relative luciferase activity of MIN6 cells cotransfected with miR-455 mimic and a luciferase reporter containing either Cpeb1-WT or Cpeb1-MUT. The data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. D: Anti-Ago2 RIP was performed in MIN6 cells transiently overexpressing miR-455, followed by qRT-PCR to detect Cpeb1 associated with Ago2 (nonspecific IgG served as NC). EG: The CPEB1 protein level in islets of len-miR-455 mice (E) (n = 5 mice/group), len-anti-miR-455 mice (F) (n = 5 mice/group), and len-anti-miR-455/ob mice (G) (n = 5 mice/group). H and I: The CPEB1 mRNA and protein levels in islets of ob/ob (H) (n = 5 mice/group) and HFD (I) (n = 5 mice/group) mice. J and K: MIN6 cells were incubated with 2.5 or 25 mmol/L glucose (J) or with 0.5 mmol/L palmitate (K) for 48 h. Then, the CPEB1 mRNA and protein levels were measured by qRT-PCR and Western blotting. L: The number of insulin-positive/KI-67+ cells were determined by immunofluorescence. Magnification, ×20; scale bar, 20 μm. M: KI-67+ β-cells were analyzed by immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001. NS, not significant.

Figure 5

miR-455 affects β-cell proliferation by targeting Cpeb1. A: Four independent miRNA target prediction algorithms were used to predict the target genes of miR-455. B: Graphic representation of the conserved miR-455 binding motif in the Cpeb1 3′-UTR of three mammalian species. Binding of the WT (top) and mutated (bottom) murine Cpeb1-3′-UTR to the miR-455 seed sequence was assessed in reporter gene experiments. C: Relative luciferase activity of MIN6 cells cotransfected with miR-455 mimic and a luciferase reporter containing either Cpeb1-WT or Cpeb1-MUT. The data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. D: Anti-Ago2 RIP was performed in MIN6 cells transiently overexpressing miR-455, followed by qRT-PCR to detect Cpeb1 associated with Ago2 (nonspecific IgG served as NC). EG: The CPEB1 protein level in islets of len-miR-455 mice (E) (n = 5 mice/group), len-anti-miR-455 mice (F) (n = 5 mice/group), and len-anti-miR-455/ob mice (G) (n = 5 mice/group). H and I: The CPEB1 mRNA and protein levels in islets of ob/ob (H) (n = 5 mice/group) and HFD (I) (n = 5 mice/group) mice. J and K: MIN6 cells were incubated with 2.5 or 25 mmol/L glucose (J) or with 0.5 mmol/L palmitate (K) for 48 h. Then, the CPEB1 mRNA and protein levels were measured by qRT-PCR and Western blotting. L: The number of insulin-positive/KI-67+ cells were determined by immunofluorescence. Magnification, ×20; scale bar, 20 μm. M: KI-67+ β-cells were analyzed by immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. Data are mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001. NS, not significant.

Close modal

To address the relevance of the miR-455-Cpeb1 interaction in the islets of obese mice, we measured the expression of Cpeb1 in pancreatic islets from ob/ob and HFD mice. As shown in Fig. 5H and I, the mRNA and protein levels of CPEB1 were significantly decreased, and similar trends were observed in MIN6 cells incubated with high glucose (Fig. 5J) or 0.5 mmol/L palmitate (Fig. 5K), indicating a targeted relationship between miR-455 and Cpeb1. We next verified whether miR-455 regulates β-cell proliferation by suppressing Cpeb1. We studied the role of Cpeb1 in β-cell proliferation, and the results showed that transfection with Cpeb1 siRNA (si-Cpeb1) markedly promoted MIN6 cell proliferation and that the effects of Cpeb1 knockdown were similar to those of miR-455 overexpression (Fig. 5L). Compared with cells transfected with si-Cpeb1, MIN6 cells transfected with si-Cpeb1 and anti-miR-455 exhibited significantly lower β-cell expansion ability (Fig. 5L and Supplementary Fig. 5G and H). Moreover, we found that len-shCpeb1–treated mice showed a higher positive KI-67 β-cell count and β-cell mass, while len-anti-miR-455 injection restored this effect (Fig. 5M and Supplementary Fig. 5I), suggesting that restoration of CPEB1 attenuated the β-cell proliferation effect of miR-455. Taken together, these findings indicate that miR-455 promotes β-cell mass expansion in a CPEB1-dependent manner.

CPEB1 Regulates Cdkn1b Expression by Promoting Its Translation Efficiency

As we previously verified, there is no targeting relationship between miR-455 and Cdkn1b, but miR-455 promotes adaptive pancreatic β-cell proliferation during obesity in a CDKN1B-dependent manner. This suggests that the regulatory effect of miR-455 on Cdkn1b is indirect. On the other hand, we confirmed that CPEB1 is a target gene of miR-455, and by controlling mRNA translation efficiency via the 3′-UTR, CPEB1 regulates many important biological processes, ranging from cell-cycle control (30) to regulation of insulin resistance (31,32). All these clues prompted us to hypothesize that CPEB1 might regulate Cdkn1b expression.

To better understand the relationship between CPEB1 and CDKN1B, we performed a bioinformatics search for target transcripts that could potentially be modulated by CPEB1. As expected, we found that Cdkn1b was one of the most promising predicted targets (Supplementary Fig. 6A). Since CPEB1 acts as a sequence-specific RNA binding protein (32), we analyzed the possible interaction between CPEB1 and Cdkn1b mRNA using RIP experiments. The RIP results revealed that the CPEB1 antibody, but not IgG, successfully pulled down Cdkn1b transcripts (Fig. 6A) and that overexpression of Cpeb1 in MIN6 cells enhanced this binding ability (Supplementary Fig. 6B). These data further confirmed that Cdkn1b mRNA is in the CPEB1 complex. In an attempt to determine the regulatory mechanisms underlying the interaction between CPEB1 and Cdkn1b, a sequence containing the WT or MUT 3′-UTR of Cdkn1b (Cdkn1b-WT and Cdkn1b-MUT, respectively) was cloned into a pGL3-control vector to construct a reporter system. When we cotransfected MIN6 cells with si-Cpeb1 and Cdkn1b-WT or Cdkn1b-MUT, we observed that oe-Cpeb1 strongly increased luciferase expression in the Cdkn1b-WT cotransfected group but not in the Cdkn1b-MUT group (Fig. 6B). These results support CPEB1 regulation of Cdkn1b mRNA through its 3′-UTR.

Figure 6

CPEB1 regulates Cdkn1b expression by promoting its translation efficiency. A: Primary islets and MIN6 cell lysates were subjected to anti-CPEB1 RIP, and Cdkn1b levels were examined via qRT-PCR. Gapdh served as a control to validate the CPEB1-Cdkn1b interaction (nonspecific IgG served as NC). B: The WT and MUT murine Cdkn1b-3′-UTR were inserted into a pGL3-control vector, and binding to Cpeb1 was assessed. Relative luciferase activity of MIN6 cells cotransfected with oe-Cpeb1 or si-Cpeb1 and a luciferase reporter containing either Cdkn1b-WT or Cdkn1b-MUT were measured. The data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. C: MIN6 cells were transfected with oe-Cpeb1 or si-Cpeb1 for 48 h, and total RNA was isolated and subjected to poly(A) tail (PAT) length assays. D and E: Western blotting (D) and qRT-PCR (E) were used to assess the CDKN1B expression level in MIN6 cells transfected with oe-Cpeb1 or si-Cpeb1. F and G: The protein (F) and mRNA (G) level of CDKN1B in islets of len-shCpeb1 mice (n = 5 mice/group). Data are mean ± SD. ***P < 0.001. NS, not significant.

Figure 6

CPEB1 regulates Cdkn1b expression by promoting its translation efficiency. A: Primary islets and MIN6 cell lysates were subjected to anti-CPEB1 RIP, and Cdkn1b levels were examined via qRT-PCR. Gapdh served as a control to validate the CPEB1-Cdkn1b interaction (nonspecific IgG served as NC). B: The WT and MUT murine Cdkn1b-3′-UTR were inserted into a pGL3-control vector, and binding to Cpeb1 was assessed. Relative luciferase activity of MIN6 cells cotransfected with oe-Cpeb1 or si-Cpeb1 and a luciferase reporter containing either Cdkn1b-WT or Cdkn1b-MUT were measured. The data are presented as the relative ratio of Renilla luciferase activity to firefly luciferase activity. C: MIN6 cells were transfected with oe-Cpeb1 or si-Cpeb1 for 48 h, and total RNA was isolated and subjected to poly(A) tail (PAT) length assays. D and E: Western blotting (D) and qRT-PCR (E) were used to assess the CDKN1B expression level in MIN6 cells transfected with oe-Cpeb1 or si-Cpeb1. F and G: The protein (F) and mRNA (G) level of CDKN1B in islets of len-shCpeb1 mice (n = 5 mice/group). Data are mean ± SD. ***P < 0.001. NS, not significant.

Close modal

To further clarify the molecular mechanisms of CPEB1 in Cdkn1b regulation, we examined whether Cdkn1b mRNA stability was affected as a consequence of CPEB1 ectopic modulation. MIN6 cells transfected with oe-Cpeb1, si-Cpeb1, or vehicle were treated with actinomycin D (10 μg/mL). Consistent with the results of Galardi et al. (33), the ectopic modulation of CPEB1 did not change the half-life of Cdkn1b mRNA, indicating that CPEB1 action is not exerted through modulation of Cdkn1b mRNA stability (Supplementary Fig. 6C). Since CPEB1 was originally identified as a sequence-specific RNA-binding protein that promotes polyadenylation-induced translation (32), we asked whether the polyadenylation status of Cdkn1b mRNA was altered by CPEB1 modulation. We performed a PCR poly(A) tail length assay on endogenous Cdkn1b mRNA in MIN6 cells transfected with oe-Cpeb1, si-Cpeb1, or vehicle. The oe-Cpeb1 enhanced the polyadenylation of Cdkn1b mRNA, while si-Cpeb1–mediated knockdown of Cpeb1 decreased polyadenylation (Fig. 6C). Because poly(A) tail lengthening is expected to result in more efficient translation, we examined whether the CDKN1B level was altered by CPEB1 ectopic modulation. As shown in Fig. 6D and E, ectopic modulation of Cpeb1 changed the protein level of CDKN1B but not the mRNA level of Cdkn1b. Analysis of CDKN1B in the islets of len-shCpeb1 mice revealed a similar trend (Fig. 6F and G). All these observations indicate that CPEB1 regulates Cdkn1b expression by promoting elongation of the poly(A) tail and translation efficiency.

The miR-455/CPEB1/CDKN1B Axis Promotes Adaptive β-Cell Expansion During Insulin Resistance

To verify that miR-455 can indirectly regulate Cdkn1b through CPEB1, we conducted a series of functional recovery experiments. Through RIP experiments, we found that the interaction between Cdkn1b and CPEB1 was significantly regulated by miR-455 (Fig. 7A and Supplementary Fig. 7A). As shown Fig. 7B, CDKN1B protein levels were decreased by Cpeb1 silencing, while anti-miR-455 restored CDKN1B expression. Analysis of CDKN1B in the islets of len-shCpeb1 or len-shCpeb1 and len-anti-miR-455 mice showed a similar trend (Fig. 7C). Next, we found that si-Cdkn1b enhanced MIN6 cell proliferation, while oe-Cpeb1 abrogated this effect (Fig. 7D and E and Supplementary Fig. 7B). Moreover, we verified that len-shCdkn1b promoted β-cell proliferation and expanded β-cell mass, while len-Cpeb1 partially restored this phenomenon (Fig. 7F and G and Supplementary Fig. 7C). These results support the role of miR-455 in promoting β-cell proliferation by inhibiting the expression of Cdkn1b via direct targeting of Cpeb1.

Figure 7

The miR-455/CPEB1/CDKN1B axis blocks β-cell proliferation during insulin resistance. A: MIN6 cells were transfected with miR-455 and anti-miR-455 for 48 h. Then, MIN6 cell lysates were subjected to anti-CPEB1 RIP, and Cdkn1b levels were examined via qRT-PCR. Gapdh served as a control to validate the CPEB1-Cdkn1b interaction (nonspecific IgG served as NC). B: The CDKN1B protein level in MIN6 cells transfected with si-Cpeb1 or si-Cpeb1 and anti-miR-455. C: The CDKN1B protein expression level in islets of len-shCpeb1 mice or len-shCpeb1 and len-anti-miR-455 mice. D and E: Insulin-positive/KI-67+ cells were counted via flow cytometry (D) and immunofluorescence (E). Magnification, ×20; scale bar, 20 μm. F: KI-67+ β-cells were analyzed via immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. G: Representative hematoxylin-eosin–stained pancreas (n = 3 mice/group). Scale bar, 200 μm. Data are mean ± SD. ***P < 0.001.

Figure 7

The miR-455/CPEB1/CDKN1B axis blocks β-cell proliferation during insulin resistance. A: MIN6 cells were transfected with miR-455 and anti-miR-455 for 48 h. Then, MIN6 cell lysates were subjected to anti-CPEB1 RIP, and Cdkn1b levels were examined via qRT-PCR. Gapdh served as a control to validate the CPEB1-Cdkn1b interaction (nonspecific IgG served as NC). B: The CDKN1B protein level in MIN6 cells transfected with si-Cpeb1 or si-Cpeb1 and anti-miR-455. C: The CDKN1B protein expression level in islets of len-shCpeb1 mice or len-shCpeb1 and len-anti-miR-455 mice. D and E: Insulin-positive/KI-67+ cells were counted via flow cytometry (D) and immunofluorescence (E). Magnification, ×20; scale bar, 20 μm. F: KI-67+ β-cells were analyzed via immunofluorescence (n = 3 mice/group). Magnification, ×20; scale bar, 20 μm. G: Representative hematoxylin-eosin–stained pancreas (n = 3 mice/group). Scale bar, 200 μm. Data are mean ± SD. ***P < 0.001.

Close modal

Obesity is frequently associated with diminished insulin sensitivity, which is normally compensated for by an expansion of the functional β-cell mass that prevents chronic hyperglycemia and the development of T2D. The molecular basis underlying compensatory β-cell mass expansion is largely unknown. In rodents, we found that β-cell mass expansion during obesity is associated with upregulated expression of miR-455, which was induced by Egr2. We also observed that oe-miR-455 increased β-cell proliferation both in vitro and in vivo. Mechanistically, we observed that the miR-455/CPEB1 pathway inhibits translation of Cdkn1b mRNA and finally leads to lower levels of CDKN1B in β-cells, in turn significantly promoting β-cell adaptive proliferation (Fig. 8). These findings point to a major role for miR-455 in compensatory β-cell mass expansion during obesity.

Figure 8

Schematic illustration of the mechanism by which the obesity-induced increase in miR-455 improves β-cell proliferation. During obesity, miR-455 is upregulated by Egr2, and miR-455 can directly target Cpeb1, suppressing CPEB1 binding to the Cdkn1b 3′-UTR, inhibiting CDKN1B expression, and finally facilitating β-cell proliferation. TSS, transcription start site.

Figure 8

Schematic illustration of the mechanism by which the obesity-induced increase in miR-455 improves β-cell proliferation. During obesity, miR-455 is upregulated by Egr2, and miR-455 can directly target Cpeb1, suppressing CPEB1 binding to the Cdkn1b 3′-UTR, inhibiting CDKN1B expression, and finally facilitating β-cell proliferation. TSS, transcription start site.

Close modal

Ob/ob mice model increased islet mass that is induced by severe insulin resistance and have been extensively studied as a model of T2D (34). In our study, we used RNA sequencing to detect differentially expressed miRNAs in the islets of ob/ob mice. Our results revealed that 85 miRNAs were significantly changed of which 65 had been identified as differentially expressed in previous research (35,36), including miR-455, miR-184, miR-375, and miR-122a. Yang et al. (37) found that miR-455 expression was increased in the serum of patients with insulin resistance. However, the mechanism of obesity-induced miR-455 upregulation and whether miR-455 can regulate β-cell function remain unclear. Therefore, we chose miR-455 for further analysis. First, to identify the possible causes of the variation in miR-455 expression in the islets of obese mice, we used a bioinformatics approach to search for putative transcription factors that could bind to the miR-455 promoter, among which Egr2 was found to be upregulated in response to palmitate treatment in primary islets and in ob/ob mouse islets. Our results are consistent with a report by Hayes et al. (38) in which the expression of Egr2 was found to be associated with T2D via genome-wide association studies. Accumulating evidence has revealed that palmitate and glucose can induce increased expression of Egr2 under insulin resistance (39,40). Here, we show that the Egr2 expression level was elevated in the pancreatic islets of obese mouse models, in MIN6 cells, and in human islets incubated with 0.5 mmol/L palmitate and 25 mmol/L glucose. Luciferase reporter, EMSA, and chromatin immunoprecipitation analyses showed that Egr2 can directly combine with the promoter of mus-miR-455 and hsa-miR-455 to induce an increase in miR-455 expression.

Here, we show that obesity induced miR-455 upregulation in islets and that the levels of miR-455 were increased in the muscle, WAT, and BAT of obese mice. miR-455 has previously been shown to activate AMPKa1 and then promote the brown adipogenic program and mitochondrial biogenesis, suggesting that miR-455 is a potential therapeutic for treating obesity (41). miR-455 expression has been evaluated in muscle with induced inflammation (42). Fang et al. (43) confirmed that miR-455 can ameliorate lipid metabolic disorders of the liver in db/db mice by inhibiting SOCS3. All these studies indicate that miR-455 may play a significant role during diabetes. However, despite this body of evidence, thus far, no studies have investigated the role of miR-455 in pancreatic β-cells. In line with previous studies showing that miR-455 plays a critical role in cell proliferation (44,45), we observed that miR-455 overexpression induced β-cell proliferation in vivo and in vitro. Furthermore, we demonstrate that β-cell–specific knockdown of miR-455 through pancreatic ductal infusion markedly aggravated HFD-induced insulin resistance resulting from a decrease in β-cell hyperplasia. Moreover, in line with islet expression analysis in insulin-resistant models, β-cell–specific knockdown of miR-455 (len-anti-miR-455) via pancreatic ductal infusion in ob/ob mice decreased the β-cell mass and KI-67 incorporation rate. The level of miR-455 was simultaneously increased in the islets of different types of obese mouse models characterized by β-cell mass expansion, suggesting a general role for miR-455 in this important compensatory mechanism. Interestingly, our in vivo experimental results revealed that insulin secretion and blood glucose level did not substantially differ between len-miR-455 and control mice. Although many studies have explored whether miRNAs play an important role in obesity and diabetes (10,46), miRNAs simply act as regulators of gene expression via posttranscriptional regulation. Thus, the function of miRNAs might not trigger pleiotropic effects. For example, Tattikota et al. (35) found that miR-184 regulates β-cell proliferation but has no effect on insulin secretion, and Jacovetti et al. (12) showed that miR-338-3p and miR-451 regulate β-cell proliferation but do not significantly affect insulin content or glucose-induced insulin secretion.

Based on a report by Shoshan et al. (47), miR-455 contributes to melanoma growth and metastasis by targeting CPEB1. In our study, we verified that miR-455 can directly target CPEB1 in β-cells, which regulate many important biological processes, ranging from cell-cycle control to insulin resistance (30,31), by controlling mRNA translation efficiency via the 3′-UTR (32,48). In agreement with these findings, we discovered that CPEB1 promotes Cdkn1b translation efficiency through elongation of the 3′-UTR of Cdkn1b mRNA. Consistent with the results of Galardi et al. (33), CPEB stimulates Cdkn1b expression by promoting elongation of the poly(A) tail and translation efficiency. Here, our results reveal that Cpeb1 overexpression facilitates Cdkn1b translation, while miR-455 restored this effect. Previous studies have shown that CDKN1B plays a negative role in the proliferation of β-cells, which is consistent with our findings that the expression level of CDKN1B is downregulated in compensatory proliferative β-cells (49,50). Altogether, these results support a role of miR-455 in promoting β-cell adaptive proliferation by inhibiting the expression of Cdkn1b via direct targeting of Cpeb1.

In summary, we demonstrate the effects of nutritional stress on miR-455, which is involved in compensatory β-cell mass expansion during obesity. Our results reveal that the miR-455/CPEB1/CDKN1B axis is required for obesity-induced adaptive pancreatic β-cell proliferation. These findings provide novel insights into the mechanism of compensatory β-cell expansion in response to increased insulin demand. Detailed knowledge of the mechanisms controlling the level and activity of miR-455 during obesity may pave the way to new therapeutic strategies, with the ultimate goal being prevention of T2D by promoting β-cell mass regeneration.

Q.H., J.M., and Yuh.L. contributed equally to this work.

This article contains supplementary material online at https://doi.org/10.2337/figshare.17189177.

Acknowledgments. The authors thank Rui Liang (Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin, China) for providing human islets.

Funding. This work was supported by the National Natural Science Foundation of China (grants 82070801 and 82100858), open fund of the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University (grant KF-GN-202103), “111” project (B16046), Priority Academic Program Development of Jiangsu Higher Education Institutions, China Postdoctoral Science Foundation (grant 2020M671661), Jiangsu Province Science Foundation for Youths (grant BK20200569), and Jiangsu Province Postdoctoral Research Fund (grant 1412000016).

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

Author Contributions. Q.H., J.M., Yuh.L., Y.Y., and Yue.L. performed the experiments, Y.P., Y.Z., L.L., and D.L. analyzed data. J.C., F.Z., and L.J. designed the project, interpreted the data, and wrote the manuscript. L.J. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Kang
T
,
Boland
BB
,
Jensen
P
, et al
.
Characterization of signaling pathways associated with pancreatic β-cell adaptive flexibility in compensation of obesity-linked diabetes in db/db mice
.
Mol Cell Proteomics
2020
;
19
:
971
993
2.
Meier
JJ
,
Breuer
TG
,
Bonadonna
RC
, et al
.
Pancreatic diabetes manifests when beta cell area declines by approximately 65% in humans
.
Diabetologia
2012
;
55
:
1346
1354
3.
Ying
W
,
Lee
YS
,
Dong
Y
, et al
.
Expansion of islet-resident macrophages leads to inflammation affecting β cell proliferation and function in obesity
.
Cell Metab
2019
;
29
:
457
474.e5
4.
Yamamoto
J
,
Imai
J
,
Izumi
T
, et al
.
Neuronal signals regulate obesity induced β-cell proliferation by FoxM1 dependent mechanism
.
Nat Commun
2017
;
8
:
1930
5.
Camastra
S
,
Manco
M
,
Mari
A
, et al
.
β-cell function in morbidly obese subjects during free living: long-term effects of weight loss
.
Diabetes
2005
;
54
:
2382
2389
6.
Donath
MY
,
Ehses
JA
,
Maedler
K
, et al
.
Mechanisms of beta-cell death in type 2 diabetes
.
Diabetes
2005
;
54
(
Suppl. 2
):
S108
S113
7.
Marselli
L
,
Suleiman
M
,
Masini
M
, et al
.
Are we overestimating the loss of beta cells in type 2 diabetes?
Diabetologia
2014
;
57
:
362
365
8.
Tyrberg
B
,
Ustinov
J
,
Otonkoski
T
,
Andersson
A
.
Stimulated endocrine cell proliferation and differentiation in transplanted human pancreatic islets: effects of the ob gene and compensatory growth of the implantation organ
.
Diabetes
2001
;
50
:
301
307
9.
Gargani
S
,
Thévenet
J
,
Yuan
JE
, et al
.
Adaptive changes of human islets to an obesogenic environment in the mouse
.
Diabetologia
2013
;
56
:
350
358
10.
Plaisance
V
,
Waeber
G
,
Regazzi
R
,
Abderrahmani
A
.
Role of microRNAs in islet beta-cell compensation and failure during diabetes
.
J Diabetes Res
2014
;
2014
:
618652
11.
Safdar
A
,
Abadi
A
,
Akhtar
M
,
Hettinga
BP
,
Tarnopolsky
MA
.
miRNA in the regulation of skeletal muscle adaptation to acute endurance exercise in C57Bl/6J male mice
.
PLoS One
2009
;
4
:
e5610
12.
Jacovetti
C
,
Abderrahmani
A
,
Parnaud
G
, et al
.
MicroRNAs contribute to compensatory β cell expansion during pregnancy and obesity
.
J Clin Invest
2012
;
122
:
3541
3551
13.
Poy
MN
,
Eliasson
L
,
Krutzfeldt
J
, et al
.
A pancreatic islet-specific microRNA regulates insulin secretion
.
Nature
2004
;
432
:
226
230
14.
Lovis
P
,
Roggli
E
,
Laybutt
DR
, et al
.
Alterations in microRNA expression contribute to fatty acid-induced pancreatic beta-cell dysfunction
.
Diabetes
2008
;
57
:
2728
2736
15.
Zhang
F
,
Ma
D
,
Zhao
W
, et al
.
Obesity-induced overexpression of miR-802 impairs insulin transcription and secretion
.
Nat Commun
2020
;
11
:
1822
16.
Rodríguez-Comas
J
,
Moreno-Asso
A
,
Moreno-Vedia
J
, et al
.
Stress-induced microRNA-708 impairs β-cell function and growth
.
Diabetes
2017
;
66
:
3029
3040
17.
Nesca
V
,
Guay
C
,
Jacovetti
C
, et al
.
Identification of particular groups of microRNAs that positively or negatively impact on beta cell function in obese models of type 2 diabetes
.
Diabetologia
2013
;
56
:
2203
2212
18.
Wang
G
,
Liang
R
,
Liu
T
, et al
.
Opposing effects of IL-1β/COX-2/PGE2 pathway loop on islets in type 2 diabetes mellitus
.
Endocr J
2019
;
66
:
691
699
19.
Zhang
FF
,
Liu
YH
,
Wang
DW
, et al
.
Obesity-induced reduced expression of the lncRNA ROIT impairs insulin transcription by downregulation of Nkx6.1 methylation
.
Diabetologia
2020
;
63
:
811
824
20.
Scharfmann
R
,
Staels
W
,
Albagli
O
.
The supply chain of human pancreatic β cell lines
.
J Clin Invest
2019
;
129
:
3511
3520
21.
Ravassard
P
,
Hazhouz
Y
,
Pechberty
S
, et al
.
A genetically engineered human pancreatic β cell line exhibiting glucose-inducible insulin secretion
.
J Clin Invest
2011
;
121
:
3589
3597
22.
Love
MI
,
Huber
W
,
Anders
S
.
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2
.
Genome Biol
2014
;
15
:
550
23.
Benjamini
Y
,
Yekutieli
D
.
The control of the false discovery rate in multiple testing under dependency
.
Ann Stat
2001
;
29
:
1165
1188
24.
Shalem
O
,
Sanjana
NE
,
Hartenian
E
, et al
.
Genome-scale CRISPR-Cas9 knockout screening in human cells
.
Science
2014
;
343
:
84
87
25.
Xiao
X
,
Guo
P
,
Prasadan
K
, et al
.
Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes
.
Nat Protoc
2014
;
9
:
2719
2724
26.
Jia
S
,
Ivanov
A
,
Blasevic
D
, et al
.
Insm1 cooperates with Neurod1 and Foxa2 to maintain mature pancreatic β-cell function
.
EMBO J
2015
;
34
:
1417
1433
27.
Kleinert
M
,
Clemmensen
C
,
Hofmann
SM
, et al
.
Animal models of obesity and diabetes mellitus
.
Nat Rev Endocrinol
2018
;
14
:
140
162
28.
Rui
L
,
Yuan
M
,
Frantz
D
,
Shoelson
S
,
White
MF
.
SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2
.
J Biol Chem
2002
;
277
:
42394
42398
29.
Cheong
MW
,
Kuo
LH
,
Cheng
YN
, et al
.
Loss of Egr-1 sensitizes pancreatic β-cells to palmitate-induced ER stress and apoptosis
.
J Mol Med (Berl)
2015
;
93
:
807
818
30.
Novoa
I
,
Gallego
J
,
Ferreira
PG
,
Mendez
R
.
Mitotic cell-cycle progression is regulated by CPEB1 and CPEB4-dependent translational control
.
Nat Cell Biol
2010
;
12
:
447
456
31.
Alexandrov
IM
,
Ivshina
M
,
Jung
DY
, et al
.
Cytoplasmic polyadenylation element binding protein deficiency stimulates PTEN and Stat3 mRNA translation and induces hepatic insulin resistance
.
PLoS Genet
2012
;
8
:
e1002457
32.
Bava
FA
,
Eliscovich
C
,
Ferreira
PG
, et al
.
CPEB1 coordinates alternative 3′-UTR formation with translational regulation
.
Nature
2013
;
495
:
121
125
33.
Galardi
S
,
Petretich
M
,
Pinna
G
, et al
.
CPEB1 restrains proliferation of glioblastoma cells through the regulation of p27(Kip1) mRNA translation
.
Sci Rep
2016
;
6
:
25219
34.
Lindström
P
.
β-cell function in obese-hyperglycemic mice [ob/ob mice]
.
Adv Exp Med Biol
2010
;
654
:
463
477
35.
Tattikota
SG
,
Rathjen
T
,
McAnulty
SJ
, et al
.
Argonaute2 mediates compensatory expansion of the pancreatic β cell
.
Cell Metab
2014
;
19
:
122
134
36.
Zhao
E
,
Keller
MP
,
Rabaglia
ME
, et al
.
Obesity and genetics regulate microRNAs in islets, liver, and adipose of diabetic mice
.
Mamm Genome
2009
;
20
:
476
485
37.
Yang
ZM
,
Chen
LH
,
Hong
M
, et al
.
Serum microRNA profiling and bioinformatics analysis of patients with type 2 diabetes mellitus in a Chinese population
.
Mol Med Rep
2017
;
15
:
2143
2153
38.
Hayes
MG
,
Pluzhnikov
A
,
Miyake
K
, et al
.
Identification of type 2 diabetes genes in Mexican Americans through genome-wide association studies
.
Diabetes
2007
;
56
:
3033
3044
39.
Bernal-Mizrachi
E
,
Wice
B
,
Inoue
H
,
Permutt
MA
.
Activation of serum response factor in the depolarization induction of Egr-1 transcription in pancreatic islet beta-cells
.
J Biol Chem
2000
;
275
:
25681
25689
40.
Chen
DL
,
Shen
DY
,
Han
CK
,
Tian
Y
.
LncRNA MEG3 aggravates palmitate-induced insulin resistance by regulating miR-185-5p/Egr2 axis in hepatic cells
.
Eur Rev Med Pharmacol Sci
2019
;
23
:
5456
5467
41.
Zhang
H
,
Guan
M
,
Townsend
KL
, et al
.
MicroRNA-455 regulates brown adipogenesis via a novel HIF1an-AMPK-PGC1α signaling network
.
EMBO Rep
2015
;
16
:
1378
1393
42.
Kinder
TB
,
Heier
CR
,
Tully
CB
, et al
.
Muscle weakness in myositis: microRNA-mediated dystrophin reduction in a myositis mouse model and human muscle biopsies
.
Arthritis Rheumatol
2020
;
72
:
1170
1183
43.
Fang
S
,
Feng
J
,
Zhang
H
, et al
.
MiR-455 targeting SOCS3 improve liver lipid disorders in diabetic mice
.
Adipocyte
2020
;
9
:
179
188
44.
Bao
C
,
Lu
Y
,
Chen
J
, et al
.
Exploring specific prognostic biomarkers in triple-negative breast cancer
.
Cell Death Dis
2019
;
10
:
807
45.
Xu
B
,
Gong
X
,
Zi
L
, et al
.
Silencing of DLEU2 suppresses pancreatic cancer cell proliferation and invasion by upregulating microRNA-455
.
Cancer Sci
2019
;
110
:
1676
1685
46.
Poy
MN
,
Hausser
J
,
Trajkovski
M
, et al
.
miR-375 maintains normal pancreatic alpha- and beta-cell mass
.
Proc Natl Acad Sci U S A
2009
;
106
:
5813
5818
47.
Shoshan
E
,
Mobley
AK
,
Braeuer
RR
, et al
.
Reduced adenosine-to-inosine miR-455-5p editing promotes melanoma growth and metastasis
.
Nat Cell Biol
2015
;
17
:
311
321
48.
Hake
LE
,
Richter
JD
.
CPEB is a specificity factor that mediates cytoplasmic polyadenylation during Xenopus oocyte maturation
.
Cell
1994
;
79
:
617
627
49.
Salisbury
RJ
,
Han
B
,
Jennings
RE
, et al
.
Altered phenotype of β-cells and other pancreatic cell lineages in patients with diffuse congenital hyperinsulinism in infancy caused by mutations in the ATP-sensitive K-channel
.
Diabetes
2015
;
64
:
3182
3188
50.
Draney
C
,
Austin
MC
,
Leifer
AH
, et al
.
HDAC1 overexpression enhances β-cell proliferation by down-regulating Cdkn1b/p27
.
Biochem J
2018
;
475
:
3997
4010
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.