1. Introduction
Diabetes is a rising concern in the world and a major cause of morbidity and mortality for the more than 400 million people living with diabetes [1, 2]. In the next 20 years, the number of people living with diabetes is expected to rise to nearly 700 million, with increasing prevalence of both of type 1 and type 2 diabetes [2]. In either type, the impotence of insulin causes dysfunction in the ability to transport glucose into cells, leading to chronically high blood sugar levels [3, 4]. Chronic diabetic complications are categorized as micro- or macrovascular, affecting capillaries and small vessels or larger vessels respectively [5]. Microvascular complications include diabetic retinopathy, diabetic nephropathy, diabetic neuropathy and diabetic cardiomyopathy (DCM), while macrovascular complications include atherosclerosis and coronary artery disease [6–8].
DCM is the microvascular cardiac complication of diabetes, and it presents with ventricular dysfunction independent of coronary atherosclerosis and hypertension [9–11]. Clinically, DCM is seen with structural and functional deviations, including left ventricular hypertrophy, cardiac fibrosis, and remodeling [12]. Owing to DCM, patients with diabetes without hypertension or coronary artery disease suffer higher rates of cardiac dysfunction than non-diabetic cohorts [10]. Microvascular pathology remains the primary contributor of DCM [5, 9, 13, 14]. At the microscopic level, DCM shows interstitial and perivascular fibrosis, cardiomyocyte hypertrophy, and cardiomyocyte death [9, 15–17]. Hyperglycemia leads to activation of deleterious pathways in endothelial cells which promote DCM. Endothelial dysfunction is signified by the prevalence of oxidative stress, an event which occurs in response to hyperglycemic damage [18–20]. In DCM, various pathways are activated in response to oxidative stress, resulting in altered signaling, and increasing the expression of VEGF, TGFβ, and NF-κB, collagen and fibronectin independent of fibroblasts [6, 21–26]. TGF-beta and NF-κB further promote the differentiation of fibroblasts to myofibroblasts [27, 28]. Additionally, endothelial cells can directly contribute to the fibrosis through a process known as endothelial-to-mesenchymal transition (EndMT), whereby the endothelial cells transdifferentiate into mesenchymal cells, contributing to the fibroblast pool [5, 29–33]. Hyperglycemic damage is also able to trigger pro-EndMT signaling [5, 34, 35].
Epigenetic changes are stable and heritable changes to gene expression without a change in the underlying genome [36]. Epigenetic modifications are central to metabolic memory and diabetic complications. Epigenetic modifications include various categories including, histone modification, DNA methylation and alteration of noncoding RNAs (ncRNAs) [37]. ncRNAs RNA molecules which are transcribed but not translated into proteins, and include long noncoding RNA, microRNA (miRNA) and circular RNA (circRNA) [38–41]. miRNA are on average 22 nucleotides long and function through binding to the 3’ UTR of target mRNA, leading to degradation in conjunction with a RNA-induced silencing complex [42]. In contrast, circRNA are covalently closed RNA molecules which function as a sponge to sequester miRNA and bind with proteins [43].
MiRNA-9-5p (miR9) is a highly conserved miRNA that has been shown to be involved in a variety of cancers and diabetic complications [44–46]. It is a molecule of interest in the heart with regards to cardiac fibrosis, and is known to be downregulated in diabetes and in response to hyperglycemia [44]. The upstream regulators of miR9 are not fully understood. We hypothesized that downregulation of miR9 in diabetes may be caused, in part, by circRNA-mediated miRNA sponging. A recent study in our lab showed an array of differentially expressed circRNAs in diabetic murine hearts, and we identified several circRNAs with predicted sponging capabilities against miR9 [47]. The current study focuses on one such circRNA, namely mmu_circRNA_012164. We investigated the glucose-induced changes in circRNA_012164 and its ability to influence miR9 in the context of diabetic heart disease.
2. Materials and methods
2.1 Animals
All animal experiments were performed in accordance with the Canadian Council on Animal Care. All experiments were approved by the University of Western Ontario Council on Animal Care Committee. Both male and female mice were used.
C57BL/6 (B6) mice were randomly divided into diabetic and control groups at 8 weeks of age. The animals were given daily intraperitoneal injections of streptozotocin (STZ) in sodium citrate buffer (pH 4.5, 50 mg/kg) for 5 consecutive days to induce diabetes. Age- and sex-matched littermates were given equal volumes of buffer only injections. Diabetes was confirmed by measuring blood glucose levels (>16.7 mmol/L) following final STZ injection. Following confirmation of diabetes, mice were weighed and checked for blood glucose, urine glucose, and urine ketones weekly. Mice were assessed for cardiac function through echocardiography. Mice were then sacrificed after 2 months using isoflurane overdose followed by cardiac puncture, and cardiac tissues were collected [44].
2.1.2 Generation of miR9 transgenic mice.
The mice used were miR9 overexpressing transgenic (m9TG) mice with EC-specific promoter previously generated in our laboratory [44]. The mice had a B6 background. Endothelial cells were further isolated from hearts of M9TG mice and endothelial specificity of miR9 overexpression was confirmed [44].
2.2 Cell culture
Mouse Cardiac Endothelial Cells (#CLU510, Cedarlane Laboratories) (MCECs) were used. Cells were incubated in a humidified incubator at 37°C with 5% CO2 in 2% DMEM (Thermo Fisher Scientific Inc) supplemented with 1% amoxicillin (Wisent Inc). The cells were serum-starved for 18 hours, then exposed to normal glucose (5 mmol/L) or high glucose (30 mmol/L) treatments of D-glucose for 48 hours. Mannitol was used as osmotic control.
MCECs were transfected with siRNA targeted circRNA_012164 (50 nM) using Lipofectamine 2000 (Invitrogen Canada Inc). Multiple anti-circRNA_012164 siRNAs were tested for efficiency, and one was chosen based on knockdown capacity (S1 Table). Scrambled siRNA was used as a control. SiRNA were designed based on complementarity to the backsplice junction of circRNA_012164. For double transfection experiments, MCECs were transfected simultaneously with a miRNA-9 antagomir [44]. Cells were incubated with OPTI-MEM and Lipofectamine 2000 for 6 hours. Cells were then recovered in DMEM with 1% serum for 6 hours, following which, they were serum starved and treated to glucose as previously described [44].
2.3 RNA isolation and qRT-PCR analysis
RNA was isolated from mouse heart tissue and MCECs using TRIzolTM reagent (Invitrogen Canada Inc, ON, Canada). qRT-PCR was performed as in previous publications in our lab [44, 45]. Linear mRNA RT was conducted at 25°C for 10 min, 37°C for 2 hr, 85°C for 5 min, and 85°C for 5 min. CircRNA RT was conducted at 25°C for 10 min, 37°C for 1.5 hr, 50°C for 30 min, and 85°C for 5 min. 2 μg of isolated RNA was used for reverse transcription. Resultant cDNA was diluted 1:10 before qPCR analyses. The reaction mixture consisted of SYBR Green Taq Ready Mix (Sigma-Aldrich), forward and reverse primers, cDNA template and H2O. Primer concentrations for PCR were in the 100–200 nM range. mRNA qPCR was conducted for 50 cycles (95°C for 10 s, 55°C for 10 s, 72°C for 15 s, 80°C for 2 s), miRNA qPCR was conducted for 50 cycles (95°C for 15 s, 60°C for 60 s, 70°C for 2 s), and circRNA qPCR was conducted for 50 cycles (95°C for 30 s, 60°C for 61 s, 72°C for 30 s, 75°C for 1 s). The data was normalized to the housekeeping gene Actb to account for variances in amount of template and reverse transcriptase efficiencies, the primer sequences are as follows (Table 1). Levels of circRNA_012164 were assessed using divergent primers designed for the backsplice junction (Table 1). Primers were additionally tested for circRNA specificity through assessment following RNAse R treatment. miR9 levels were assessed with TaqMan miRNA assay (Ambion Inc) following miRNA column extraction (Bio Basic) and normalized to U6 snRNA, in accordance with previous protocol [44].
[Figure omitted. See PDF.]
2.4 Protein isolation and analysis
The tissues were lysed and ultrasonicated in RIPA buffer (MilliporeSigma) after harvest. Total protein was collected, and the concentration was measured by using BCA kit (Thermo Fisher Scientific Inc). ELISAs for mouse FN1 (Boster Bio) and COL1A1 (Novus Biologicals) were performed [44].
2.5 Histology
Mouse heart tissues were collected, fixed in 10% formalin, embedded in paraffin, and cut to 5 μm thick sections on positively charged slides. The sections were deparaffinized in xylene and stained with hematoxylin and eosin as well as Masson’s trichrome stain [44].
2.6 Statistical analysis
The data are expressed as mean ± standard error of the mean. Statistical significances were analyzed by Student’s t-test or ANOVA, followed by Tukey test for multiple comparisons. Differences were considered to be statistically significant at values of p < 0.05. Statistical analyses and graphs were generated using GraphPad Prism version 8.0.2 for Windows (GraphPad Software).
3. Results
3.1 circRNA_012164 and microRNA-9-5p levels are altered in the heart in diabetes
Diabetic animals showed evidence of dysmetabolism as expected. For both blood glucose and body weight, diabetic mice of both the B6 and m9TG lines showed hyperglycemia and reduced body weight when compared to non-diabetic (Table 2). They also demonstrated polyuria (not shown). Diabetic m9TG mice showed similar loss of body weight and hyperglycemia as diabetic wildtype mice (Table 2).
[Figure omitted. See PDF.]
To validate previous data from our laboratory, miR9 levels were assessed. We confirmed that miR9 was significantly inhibited by diabetes in the murine heart (Fig 1A). In addition, CircRNA_012164 levels were also elevated in the hearts of diabetic mice (Fig 1B), in accordance with previous literature [47]. Changes in ncRNA expression were consistent across heart tissues of both male and female mice (S1 Fig).
[Figure omitted. See PDF.]
Assessment of A) microRNA-9 and B) circRNA_01216 levels showed decreased expression of miR-9 and increased expression of circRNA_012164 in male diabetic mice compared to age- and sex-matched non-diabetic controls. Female mice showed similar changes (S1 Fig). (Data are expressed as ± SEM and normalized to U6 or Actb. * = significantly different. n = 7/group).
3.2 microRNA-9-5p overexpression prevents diabetes-induced cardiac fibrosis
After validating glucose-induced changes in our ncRNAs of interest, we then proceeded to directly examine the role of miR9 in the heart in diabetes. To this extent we used our custom-generated endothelial-specific miR9 overexpressing (m9TG) mice. No phenotypic differences were observed in m9TG mice compared to B6 via echocardiographic analysis, however, B6 diabetic mice showed a pseudonormal E/A ratio when compared to the normal E/A ratios of other groups (Fig 2). The same changes were observed in female mice (S2 Fig).
[Figure omitted. See PDF.]
Echocardiographic analysis showed significant reduction of the A) E/A ratio of wild-type diabetic (B6-D) compared to wild-type non-diabetic (B6-C) mice. This reduction was prevented in endothelial specific miR-9 overexpressing transgenic mice. No phenotypic changes were observed in A) E/A ratio, B) fractional shortening, or C) ejection fraction between B6-C and either miR-9 transgenic groups. Representative echocardiography tracings are shown: (top panel) end-systolic diameter and end-diastolic diameter, (middle panel) pulsed wave Doppler recording of mitral inflow velocity, and (bottom panel) tissue Doppler imaging of the LV posterior wall of D) B6-C, E) B6-D, F) miR9-C, and G) miR9-D. Female mice showed similar changes (S2 Fig). (Data are expressed as mean ± SEM. * = significantly different from B6-C. n = 8/group).
Gene expression associated with the molecules altered in fibrosis were examined in murine hearts from both B6 and m9TG mice, encompassing both diabetic and non-diabetic conditions. Trichrome staining of murine heart tissue indicated that diabetic B6 mice showed interstitial fibrosis, whereas the age- and sex- matched control diabetic m9TG mice did not (Fig 3A–3D). This analysis further included assessment of mRNAs for Col1a1 and Fn1, genes encoding extracellular matrix proteins, and S100a4, a gene that serves as a fibroblast activation marker (Fig 3E–3G). To validate that mRNA expression levels corresponded with observable changes in phenotype, we conducted protein analyses for two key ECM proteins, COL1A1 and FN1 (Fig 3H and 3I). All the genes assessed were significantly elevated in B6 diabetic mouse hearts. These changes indicate diabetes associated cardiac damage. In addition, diabetic animals showed presence of focal myocardial fibrosis. All such alterations were effectively prevented in diabetic m9TG mice (Fig 3).
[Figure omitted. See PDF.]
Masson’s Trichrome staining showed cardiac fibrosis in B) wild-type diabetic (B6-D) compared to A) wild-type non-diabetic (B6-C) hearts, as indicated by arrows. Cardiac fibrosis was prevented in the D) diabetic microRNA-9 overexpressing (endothelial specific) transgenic mice (miR9-D). No differences were observed between A) B6-C and C) miR-9 transgenic non-diabetic (miR9-C) mice. mRNA expressions of E) Col1a1, F) Fn1, and G) S100a4 were upregulated in B6-D compared to B6-C hearts. These changes were prevented in miR-9 transgenic mice. mRNA expressions were normalized to Actb. Protein levels of H) COL1A1 and I) FN1 were similarly changed in B6-D and miR9-D groups. No differences were observed based on sex differences (S1 Fig). (Data are expressed as mean ± SEM. * = statistically significant. n = 6–8/group for mRNA analyses, n = 3/group for protein analyses).
3.3 Knockdown of circRNA_012164 prevented glucose-induced changes in cardiac endothelial cells
After showing the protective effects of miR-9 against DCM-related fibrotic changes in vivo, we continued the experiments in isolated mouse cardiac endothelial cells (MCECs) to gain mechanistic insights specific to endothelial cells and endothelial dysfunction. In response to treatment with 30 mM D-glucose, MCECs showed significant elevation in the levels of circRNA_012164 as well as a reduction in miR9 levels corresponding to previous literature and our in vivo studies [44] (Fig 4A and 4B). No such changes were observed with osmotic control using mannitol (not shown). Similar to our findings in vivo, the mRNA expressions of ECM genes Col1a1 and Fn1 were significantly upregulated in response to high glucose (Fig 4D and 4E). Similarly, the levels of Acta2 and S100a4, recognized markers of mesenchymal cells and fibroblasts respectively, also demonstrated an upswing (Fig 4C and 4F). Particularly noteworthy was the heightened expression of Acta2, an indication that endothelial cells had initiated the transition toward a mesenchymal phenotype. Knockdown of circRNA_012164 proved to be effective in rescuing aforementioned changes in markers of fibrosis and indicates possible EndMT, while also restoring miR9 levels to those seen in normoglycemic conditions (Fig 4). Interestingly, aside from its protective role in hyperglycemia, circRNA_012164 knockdown had no significant impact on basal levels of any genes, except for Fn1 (Fig 4E).
[Figure omitted. See PDF.]
High glucose (30 mM, HG) treatment of mouse cardiac endothelial cells resulted in downregulation of A) miR-9 and upregulation of B) circRNA_012164 along with the downstream genes C) Acta2 D) Col1a1, E) S100a4, and F) Fn1. siRNA targeted to circRNA_012164 (si-circ) protected cells against HG-induced changes. Data normalized to U6 or Actb where applicable. (Data are expressed as mean ± SEM. * = p < 0.05, n = 5–6/group.) NG = normal glucose, 5 mM; scr = scrambled oligonucleotides.
3.4 Inhibition of microRNA-9 reverts the protective effects of circRNA_012164 knockdown
Having shown the effects of circRNA_012164 knockdown on miR-9 levels and the development of endothelial fibrogenesis and mesenchymal transition, we used a rescue strategy to establish a direct regulatory pathway. To this extent, MCECs were co-transfected with anti- circRNA_012164 siRNA and an antagomir targeting miR9. Co-transfection with both si-circ012164 and miR9 antagomir resulted in a complete nullification of all protective effects of circRNA_012164 knockdown. Co-transfected cells showed significant upregulation of our genes of interest, on par with cells transfected with scrambled oligonucleotides (Fig 5).
[Figure omitted. See PDF.]
High glucose (30 mM, HG) treatment of mouse cardiac endothelial cells resulted in upregulation of A) Acta2 B) Col1a1, C) S100a4, and D) Fn1. Knockdown of circRNA_012164 (si-circ) prevented (HG) induced gene expression changes. Additionally inhibiting microRNA-9 (anti-m9) removed protective effect of si-circ and restored the HG phenotype. Data normalized to Actb. (Data are expressed as mean ± SEM. * = p < 0.05, n = 5–6/group.) NG = normal glucose, 5 mM; scr = scrambled oligonucleotides.
4. Discussion
The aim of this study was to investigate novel epigenetic mechanisms to develop a better understanding of the pathogenic processes involved in diabetic cardiomyopathy. DCM is an independent risk factor for heart failure in diabetes and manifests clinically as a loss of contractility with cardiac fibrosis and remodeling [16]. Previous research done in our lab has found the involvement of multiple epigenetic mechanisms, including histone acetylation, long-noncoding RNA, and other miRNAs in the pathogenesis of DCM [44, 48–51]. We had also previously identified several differentially expressed circRNA in the heart in diabetes and their predicted miRNA targets [47].
In this study, we focused on circRNA_012164, which was upregulated in the heart in diabetes. CircRNA_012164 was predicted to regulate miR9, which has previously been identified as a regulator of diabetic complications in multiple organs, including DCM [47]. Our investigation established regulatory pathways in various models, both in in vitro and in vivo, using both genetically modified and chronically diabetic mice. Overall, our results indicate the existence of the circRNA_012164/miR9 regulatory axis in the pathogenesis of cardiac fibrosis in DCM. Cardiac fibrosis manifests as increased production of ECM proteins such as collagen type I and fibronectin [52]. ECs are capable of both activating fibroblasts through signaling derangement and differentiating into fibroblasts through EndMT, a process by which transcription derangement leads to ECs taking on a mesenchymal phenotype [30].
Levels of miR9 were found to be downregulated in the heart in diabetes, consistent with previous studies [44]. In addition, an increase in transcripts of genes encoding the ECM proteins collagen type 1 (Col1a1) and fibronectin (Fn1), as well as an increase in the fibroblast marker, S100a4. We further show the potency of miR9’s regulatory role using an EC-specific miR9 overexpressing transgenic mouse model. Through endothelial-specific expression of miR9, levels of COL1A1 and FN1 were both significantly reduced in diabetes and restored to baseline non-diabetic. In addition, S100a4 was also significantly reduced and restored to baseline. This is consistent with previous studies that miR9 has an effect on mediating and preventing the development of cardiac fibrosis in response to diabetes [44]. While miR9 has been quantified previously as being a regulator of genes contributing to various diabetic complications through interactions with proteins and other ncRNA, little is known about the mechanisms behind regulation of miR9 [44, 45]. We have previously shown that lncRNA ZFAS1 regulates miR9 through induction of trimethylation of H3K27 via coordination of the polycomb repressive complex 2 [44]. CircRNA_012164 in this case affects miR9 levels at the post-transcriptional level.
CircRNAs are known to regulate miRNA through competitive endogenous binding through sequence complementarity [53]. In diabetic retinopathy, circRNA_0005015 levels were found to be upregulated and facilitated endothelial angiogenesis by promoting EC proliferation, migration and tube formation through inhibition of miR-519d-3p [53]. In DCM, circRNA_010567 was found to be upregulated in diabetic mouse myocardium and in cardiac fibroblasts, and promoted fibrosis through interactions with miR-141 [54]. Our study identified circRNA_012164 as an upstream regulator of miR9. CircRNA_012164 was found to be upregulated in diabetes, consistent with previous findings [47].
Knockdown of circRNA_012164 in normal glucose conditions did not affect baseline expression of most genes, indicating that the effect observed in high glucose conditions could be attributed to glucose-dependent upstream regulators of circRNA_012164. As circRNAs function through sponging, there may be a co-inhibitory effect between circRNA_012164 and miR9. Double knockdown of both circRNA_012164 and miR9 shows that the effect of circRNA_012164 knockdown is tied to miR9. The findings indicate a circRNA_012164/miR9 regulatory axis, and that the profibrotic effect of circRNA_012164 is in part mediated through miR9 inhibition. Regulation of circRNA_012164 and indeed other circRNA have yet to be fully researched.
CircRNAs may be regulated at the transcriptional level through regulation of their parental gene. While the regulators of circRNA_012164 expression are unknown, its parent gene, Hist1h1c, codes for the Histone H1.2 protein and we may identify regulators from Hist1h1c [47]. Hist1h1c has been found to be upregulated and promoting apoptosis in diabetic retinopathy, further suggesting that high glucose may promote expression of His1h1c and subsequently, circRNA_012164 [55]. An increasing body of evidence suggests that circRNAs also reciprocally regulates parental gene expression [56, 57]. In this capacity, circRNAs may interface with transcriptional machinery proteins and in the post-transcriptional splicing stage [58, 59]. No evidence exists of this for circRNA_012164, as such this remains an area for further exploration.
Levels of circRNA_012164 changed in whole cardiac tissue as well as in cardiac endothelial cells. This suggests that other cells in the heart may also express circRNA_012164. Previous studies have shown that miR9 is downregulated in cardiomyocytes and in diabetic hearts, and that miR9 inhibits cardiomyocyte death [60]. Should circRNA_012164 be expressed in other cardiac cells as well, it would indicate a potential future exploration into the role of circRNA_012164 in the regulation of cardiomyocytes and resident cardiac fibroblasts. CircRNA_012164 and miR9 may also function in cardiomyocytes and cardiac fibroblasts through cell-cell interactions with endothelial cells. Gap junctions between endothelial cells and cardiomyocytes and between endothelial cells and cardiac fibroblasts may allow for the transport of ncRNA through connexons [61, 62]. Finally, extracellular vesicles formed from endothelial cells may transport ncRNA to cells without requiring proximity as shown in various studies, they have been indicated as playing a role in endothelial dysfunction as well as other diseases of vascular damage [63, 64]. In diabetes, miR-9-3p has been found to be transferred to retinal endothelial cells in diabetic retinopathy; similar occurrences in DCM require further investigation [65].
In addition, a previous study from our lab have shown that several other circRNA with diabetically altered expression that bind to various miRNA of interest, including miR9 [47]. Other upregulated circRNA, like circRNA_39888, bind to miR-200b, another miRNA of interest in diabetes [47, 50]. Investigation as to the biologic significance of these differentially expressed circRNA may further illustrate a more comprehensive network of interlinked regulatory molecules that contribute to a disease process as complicated as DCM.
In conclusion, we have established the role of circRNA_012164 and its interactions with miR9 in the context of DCM. We have found that circRNA_012164 is upregulated in diabetic murine hearts and that it promotes endothelial dysfunction and fibrogenesis in cardiac endothelial cells. Further investigation into circRNA_012164 upstream regulation as well as other circRNA targeted to miR9 should be done to gain better insight into the role of ncRNA in DCM.
Supporting information
S1 Fig. MiR9 and circRNA_012164 expression changes in hearts of diabetic female mice show changes comparable to male mice.
https://doi.org/10.1371/journal.pone.0302772.s001
(TIF)
S2 Fig.
A) E/A ration, B) fractional shortening percent and C) ejection fraction percent in female mouse echocardiography. No differences were shown between male and female mice.
https://doi.org/10.1371/journal.pone.0302772.s002
(TIF)
S1 Table. Sequence of siRNA targeted to the backsplice junction of circRNA_012164.
https://doi.org/10.1371/journal.pone.0302772.s003
(TIF)
Citation: Wang H, Wang EZR, Feng B, Chakrabarti S (2024) CircRNA_012164/MicroRNA-9-5p axis mediates cardiac fibrosis in diabetic cardiomyopathy. PLoS ONE 19(7): e0302772. https://doi.org/10.1371/journal.pone.0302772
About the Authors:
Honglin Wang
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Writing – original draft, Writing – review & editing
Affiliation: Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
Eric Zi Rui Wang
Roles: Validation, Writing – review & editing
Affiliation: Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
ORICD: https://orcid.org/0000-0001-5140-5567
Biao Feng
Roles: Conceptualization, Methodology, Project administration, Supervision
E-mail: [email protected] (SC); [email protected] (BF)
Affiliation: Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
Subrata Chakrabarti
Roles: Funding acquisition, Supervision, Writing – review & editing
E-mail: [email protected] (SC); [email protected] (BF)
Affiliation: Department of Pathology and Laboratory Medicine, University of Western Ontario, London, Ontario, Canada
ORICD: https://orcid.org/0000-0002-0208-1401
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
1. Lin X, Xu Y, Pan X, Xu J, Ding Y, Sun X, et al. Global, regional, and national burden and trend of diabetes in 195 countries and territories: an analysis from 1990 to 2025. Sci Rep. 2020 Sep 8;10(1):14790. pmid:32901098
2. Cho NH, Shaw JE, Karuranga S, Huang Y, Fernandes JD da R, Ohlrogge AW, et al. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Research and Clinical Practice. 2018 Apr 1;138:271–81. pmid:29496507
3. Alam F, Islam MA, Khalil MI, Gan SH. Metabolic Control of Type 2 Diabetes by Targeting the GLUT4 Glucose Transporter: Intervention Approaches. Curr Pharm Des. 2016;22(20):3034–49. pmid:26951104
4. Vargas E, Podder V, Carrillo Sepulveda MA. Physiology, Glucose Transporter Type 4. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Jul 8]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK537322/
5. Wang E, Wang H, Chakrabarti S. Endothelial-to-mesenchymal transition: An underappreciated mediator of diabetic complications. Frontiers in Endocrinology [Internet]. 2023 [cited 2023 Jul 8];14. Available from: https://www.frontiersin.org/articles/10.3389/fendo.2023.1050540 pmid:36777351
6. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005 Jun;54(6):1615–25. pmid:15919781
7. Chan L, Terashima T, Fujimiya M, Kojima H. Chronic diabetic complications: the body’s adaptive response to hyperglycemia gone awry? Trans Am Clin Climatol Assoc. 2006;117:341–51; discussion 351–352. pmid:18528485
8. Fowler MJ. Microvascular and Macrovascular Complications of Diabetes. Clinical Diabetes. 2011 Jul 1;29(3):116–22.
9. Jia G, Whaley-Connell A, Sowers JR. Diabetic cardiomyopathy: a hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia. 2018 Jan;61(1):21–8. pmid:28776083
10. Avogaro A, Azzolina D, Fadini GP, Baldi I. Incidence of heart failure in patients with type 1 diabetes: a systematic review of observational studies. J Endocrinol Invest. 2021 Apr;44(4):745–53. pmid:32734319
11. Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW, Grishman A. New type of cardiomyopathy associated with diabetic glomerulosclerosis. American Journal of Cardiology. 1972 Nov 8;30(6):595–602. pmid:4263660
12. Jia G, Hill MA, Sowers JR. Diabetic Cardiomyopathy. Circulation Research. 2018 Feb 16;122(4):624–38.
13. Gamrat A, Surdacki MA, Chyrchel B, Surdacki A. Endothelial Dysfunction: A Contributor to Adverse Cardiovascular Remodeling and Heart Failure Development in Type 2 Diabetes beyond Accelerated Atherogenesis. J Clin Med. 2020 Jul 3;9(7):2090. pmid:32635218
14. Hinkel R, Howe A, Renner S, Ng J, Lee S, Klett K, et al. Diabetes Mellitus–Induced Microvascular Destabilization in the Myocardium. Journal of the American College of Cardiology. 2017 Jan 17;69(2):131–43. pmid:28081822
15. Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018 Feb 16;122(4):624–38. pmid:29449364
16. Athithan L, Gulsin GS, McCann GP, Levelt E. Diabetic cardiomyopathy: Pathophysiology, theories and evidence to date. World J Diabetes. 2019 Oct 15;10(10):490–510. pmid:31641426
17. Evidence for Cardiomyopathy in Familial Diabetes Mellitus ‐ PMC [Internet]. [cited 2023 Jul 10]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC372437/
18. Premer C, Kanelidis AJ, Hare JM, Schulman IH. Rethinking Endothelial Dysfunction as a Crucial Target in Fighting Heart Failure. Mayo Clin Proc Innov Qual Outcomes. 2019 Feb 26;3(1):1–13. pmid:30899903
19. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010 Oct 29;107(9):1058–70. pmid:21030723
20. Asmat U, Abad K, Ismail K. Diabetes mellitus and oxidative stress—A concise review. Saudi Pharm J. 2016 Sep;24(5):547–53. pmid:27752226
21. Barrett EJ, Liu Z. The Endothelial Cell: an “Early Responder” in the Development of Insulin Resistance. Rev Endocr Metab Disord. 2013 Mar;14(1):21–7. pmid:23306779
22. Zhang J, Wang YJ, Wang X, Xu L, Yang XC, Zhao WS. PKC-Mediated Endothelin-1 Expression in Endothelial Cell Promotes Macrophage Activation in Atherogenesis. Am J Hypertens. 2019 Aug 14;32(9):880–9. pmid:31111864
23. Yasuda Y, Nakamura J, Hamada Y, Nakayama M, Chaya S, Naruse K, et al. Role of PKC and TGF-beta receptor in glucose-induced proliferation of smooth muscle cells. Biochem Biophys Res Commun. 2001 Feb 16;281(1):71–7. pmid:11178962
24. Pan D, Xu L, Guo M. The role of protein kinase C in diabetic microvascular complications. Front Endocrinol (Lausanne). 2022 Aug 17;13:973058. pmid:36060954
25. Aveleira CA, Lin CM, Abcouwer SF, Ambrósio AF, Antonetti DA. TNF-α Signals Through PKCζ/NF-κB to Alter the Tight Junction Complex and Increase Retinal Endothelial Cell Permeability. Diabetes. 2010 Nov;59(11):2872–82.
26. Lee HB, Yu MR, Song JS, Ha H. Reactive oxygen species amplify protein kinase C signaling in high glucose-induced fibronectin expression by human peritoneal mesothelial cells. Kidney International. 2004 Apr 1;65(4):1170–9. pmid:15086456
27. Saxena A, Dobaczewski M, Rai V, Haque Z, Chen W, Li N, et al. Regulatory T cells are recruited in the infarcted mouse myocardium and may modulate fibroblast phenotype and function. Am J Physiol Heart Circ Physiol. 2014 Oct 15;307(8):H1233–42. pmid:25128167
28. Waitkus MS, Harris DP, DiCorleto PE. Mechanisms of Endothelial Activation. In: Mackay IR, Rose NR, Diamond B, Davidson A, editors. Encyclopedia of Medical Immunology: Autoimmune Diseases [Internet]. New York, NY: Springer; 2014 [cited 2023 Jul 10]. p. 715–24. Available from: https://doi.org/10.1007/978-0-387-84828-0_183
29. Cooley BC, Nevado J, Mellad J, Yang D, St. Hilaire C, Negro A, et al. TGF-β Signaling Mediates Endothelial-to-Mesenchymal Transition (EndMT) During Vein Graft Remodeling. Science Translational Medicine. 2014 Mar 12;6(227):227ra34-227ra34.
30. Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007 Aug;13(8):952–61. pmid:17660828
31. Bischoff J. Endothelial-to-Mesenchymal Transition. Circ Res. 2019 Apr 12;124(8):1163–5. pmid:30973806
32. Li J, Qu X, Bertram JF. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol. 2009 Oct;175(4):1380–8. pmid:19729486
33. Krizbai IA, Gasparics Á, Nagyőszi P, Fazakas C, Molnár J, Wilhelm I, et al. Endothelial-Mesenchymal Transition of Brain Endothelial Cells: Possible Role during Metastatic Extravasation. PLOS ONE. 2015 Mar 5;10(3):e0119655. pmid:25742314
34. Piera-Velazquez S, Jimenez SA. Endothelial to Mesenchymal Transition: Role in Physiology and in the Pathogenesis of Human Diseases. Physiol Rev. 2019 Apr 1;99(2):1281–324. pmid:30864875
35. Yu CH, Suriguga null, Gong M, Liu WJ, Cui NX, Wang Y, et al. High glucose induced endothelial to mesenchymal transition in human umbilical vein endothelial cell. Exp Mol Pathol. 2017 Jun;102(3):377–83. pmid:28347704
36. Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003 Mar;33 Suppl:245–54. pmid:12610534
37. Gibney ER, Nolan CM. Epigenetics and gene expression. Heredity. 2010 Jul;105(1):4–13. pmid:20461105
38. Yao Q, Chen Y, Zhou X. The roles of microRNAs in epigenetic regulation. Curr Opin Chem Biol. 2019 Aug;51:11–7. pmid:30825741
39. Ponting CP, Oliver PL, Reik W. Evolution and Functions of Long Noncoding RNAs. Cell. 2009 Feb 20;136(4):629–41. pmid:19239885
40. Holoch D, Moazed D. RNA-mediated epigenetic regulation of gene expression. Nat Rev Genet. 2015 Feb;16(2):71–84. pmid:25554358
41. Zhang M, Xu K, Fu L, Wang Q, Chang Z, Zou H, et al. Revealing Epigenetic Factors of circRNA Expression by Machine Learning in Various Cellular Contexts. iScience. 2020 Nov 24;23(12):101842. pmid:33319172
42. MacFarlane LA, Murphy PR. MicroRNA: Biogenesis, Function and Role in Cancer. Curr Genomics. 2010 Nov;11(7):537–61. pmid:21532838
43. Qin T, Li J, Zhang KQ. Structure, Regulation, and Function of Linear and Circular Long Non-Coding RNAs. Frontiers in Genetics [Internet]. 2020 [cited 2023 Sep 13];11. Available from: https://www.frontiersin.org/articles/10.3389/fgene.2020.00150 pmid:32194627
44. Feng B, Liu J, Wang E, Su Z, Chakrabarti S. Endothelial derived miRNA-9 mediated cardiac fibrosis in diabetes and its regulation by ZFAS1. PLoS One. 2022 Oct 14;17(10):e0276076. pmid:36240130
45. Wang E, Feng B, Chakrabarti S. MicroRNA 9 Is a Regulator of Endothelial to Mesenchymal Transition in Diabetic Retinopathy. Invest Ophthalmol Vis Sci. 2023 Jun 6;64(7):13. pmid:37279396
46. Han D, Li J, Wang H, Su X, Hou J, Gu Y, et al. Circular RNA circMTO1 acts as the sponge of microRNA-9 to suppress hepatocellular carcinoma progression. Hepatology. 2017 Oct;66(4):1151–64. pmid:28520103
47. Patil NS, Feng B, Su Z, Castellani CA, Chakrabarti S. Circular RNA mediated gene regulation in chronic diabetic complications. Sci Rep. 2021 Dec 9;11(1):23766. pmid:34887449
48. Biswas S, Chakrabarti S. Increased Extracellular Matrix Protein Production in Chronic Diabetic Complications: Implications of Non-Coding RNAs. Noncoding RNA. 2019 Mar 22;5(1):E30. pmid:30909482
49. Chen S, Feng B, Thomas AA, Chakrabarti S. miR-146a regulates glucose induced upregulation of inflammatory cytokines extracellular matrix proteins in the retina and kidney in diabetes. PLoS One. 2017 Mar 16;12(3):e0173918. pmid:28301595
50. Feng B, Cao Y, Chen S, Chu X, Chu Y, Chakrabarti S. miR-200b Mediates Endothelial-to-Mesenchymal Transition in Diabetic Cardiomyopathy. Diabetes. 2016 Mar 1;65(3):768–79. pmid:26718496
51. Feng B, Chen S, Gordon AD, Chakrabarti S. miR-146a mediates inflammatory changes and fibrosis in the heart in diabetes. J Mol Cell Cardiol. 2017 Apr;105:70–6. pmid:28279663
52. Kong P, Christia P, Frangogiannis NG. The Pathogenesis of Cardiac Fibrosis. Cell Mol Life Sci. 2014 Feb;71(4):549–74. pmid:23649149
53. Zhang SJ, Chen X, Li CP, Li XM, Liu C, Liu BH, et al. Identification and Characterization of Circular RNAs as a New Class of Putative Biomarkers in Diabetes Retinopathy. Investigative Ophthalmology & Visual Science. 2017 Dec 29;58(14):6500–9. pmid:29288268
54. Zhou B, Yu JW. A novel identified circular RNA, circRNA_010567, promotes myocardial fibrosis via suppressing miR-141 by targeting TGF-β1. Biochemical and Biophysical Research Communications. 2017 Jun 10;487(4):769–75.
55. Wang W, Wang Q, Wan D, Sun Y, Wang L, Chen H, et al. Histone HIST1H1C/H1.2 regulates autophagy in the development of diabetic retinopathy. Autophagy. 2017 Mar 2;13(5):941–54. pmid:28409999
56. Shao T, Pan Y hong, Xiong X dong. Circular RNA: an important player with multiple facets to regulate its parental gene expression. Mol Ther Nucleic Acids. 2020 Nov 17;23:369–76. pmid:33425494
57. Wang H, Gao X, Yu S, Wang W, Liu G, Jiang X, et al. Circular RNAs regulate parental gene expression: A new direction for molecular oncology research. Front Oncol. 2022 Aug 25;12:947775. pmid:36091137
58. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 2015 Mar;22(3):256–64. pmid:25664725
59. Conn VM, Hugouvieux V, Nayak A, Conos SA, Capovilla G, Cildir G, et al. A circRNA from SEPALLATA3 regulates splicing of its cognate mRNA through R-loop formation. Nat Plants. 2017 Apr 18;3:17053. pmid:28418376
60. Jeyabal P, Thandavarayan RA, Joladarashi D, Babu SS, Krishnamurthy S, Bhimaraj A, et al. MicroRNA-9 inhibits hyperglycemia induced cardiac pyroptosis in human ventricular cardiomyocytes by targeting ELAVL1. Biochem Biophys Res Commun. 2016 Mar 18;471(4):423–9.
61. Talman V, Kivelä R. Cardiomyocyte—Endothelial Cell Interactions in Cardiac Remodeling and Regeneration. Front Cardiovasc Med [Internet]. 2018 Jul 26 [cited 2024 Mar 19];5. Available from: https://www.frontiersin.org/articles/10.3389/fcvm.2018.00101 pmid:30175102
62. Colliva A, Braga L, Giacca M, Zacchigna S. Endothelial cell–cardiomyocyte crosstalk in heart development and disease. J Physiol. 2020 Jul 15;598(14):2923–39. pmid:30816576
63. Sima AA, Nathaniel V, Prashar A, Bril V, Greene DA. Endoneurial microvessels in human diabetic neuropathy. Endothelial cell dysjunction and lack of treatment effect by aldose reductase inhibitor. Diabetes. 1991 Sep;40(9):1090–9. pmid:1936616
64. Schiano C, Balbi C, de Nigris F, Napoli C. Basic Pathogenic Mechanisms and Epigenetic Players Promoted by Extracellular Vesicles in Vascular Damage. Int J Mol Sci. 2023 Apr 19;24(8):7509. pmid:37108672
65. Liu Y, Yang Q, Fu H, Wang J, Yuan S, Li X, et al. Müller glia-derived exosomal miR-9-3p promotes angiogenesis by restricting sphingosine-1-phosphate receptor S1P1 in diabetic retinopathy. Mol Ther Nucleic Acids. 2022 Mar 8;27:491–504.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 Wang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Noncoding RNAs play a part in many chronic diseases and interact with each other to regulate gene expression. MicroRNA-9-5p (miR9) has been thought to be a potential inhibitor of diabetic cardiomyopathy. Here we examined the role of miR9 in regulating cardiac fibrosis in the context of diabetic cardiomyopathy. We further expanded our studies through investigation of a regulatory circularRNA, circRNA_012164, on the action of miR9. We showed at both the in vivo and in vitro level that glucose induced downregulation of miR9 and upregulation of circRNA_012164 resulted in the subsequent upregulation of downstream fibrotic genes. Further, knockdown of circRNA_012164 shows protective effects in cardiac endothelial cells and reverses increased transcription of genes associated with fibrosis and fibroblast proliferation through a regulatory axis with miR9. This study presents a novel regulatory axis involving noncoding RNA that is evidently important in the development of cardiac fibrosis in diabetic cardiomyopathy.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer