MiR-18b suppresses high-glucose-induced proliferation in HRECs by targeting IGF-1/IGF1R signaling pathways

https://doi.org/10.1016/j.biocel.2016.02.002Get rights and content

Highlights

  • High glucose promotes HRECs proliferation by downregulating miR-18b.

  • MiR-18b directly target 3′ UTR of human IGF-1 in vitro.

  • Activation of IGF-1/IGF1R pathway mediates the effect of miR-18b on proliferation and VEGF production.

  • Overexpression of miR-18b might offer novel therapeutic options for DR.

Abstract

MicroRNAs (miRNAs) are important for the proliferation of endothelial cells and have been shown to be involved in diabetic retinopathy (DR). In previous study, we found that miRNAs might play a critical role in hyperglycemia-induced endothelial cell proliferation based on miRNA expression profiling. Here, the roles of microRNA-18b (miR-18b) in the proliferation of human retinal endothelial cells (HRECs) were investigated in an in vitro model of HRECs grown in high glucose. We identified that levels of miR-18b were decreased in high-glucose-induced HRECs, compared with those in cells incubated in normal glucose. However, the reduction of miR-18b up-regulated vascular endothelial growth factor (VEGF) secretion and promoted effects on in vitro proliferation of HRECs. Mechanistically, insulin growth factor-1 (IGF-1) was identified as a target of miR-18b. IGF-1 simulation could antagonize the effect induced by miR-18b up-regulation, promoting cell proliferation and increasing VEGF production. In contrast, the opposite results were observed with silencing IGF-1, which was consistent with the effects of miR-18b overexpression. MiR-18b exerted its function on VEGF synthesis and cell proliferation by suppressing the IGF-1/insulin growth factor-1 receptor (IGF1R) pathway, consequently inhibiting the downstream phosphorylation of Akt, MEK, and ERK. Hence, this may provide a new insight into understanding the mechanism of DR pathogenesis, as well as a potential therapeutic target for proliferative DR.

Introduction

Diabetic retinopathy (DR) is the most common micro-vascular complication of diabetes and one of the main reasons of blindness in adults in developed countries (Fong et al., 2004, Moss et al., 1998). Retinal changes in diabetes are thought to be initiated by sustained hyperglycemia, which has been recognized as a major risk factor responsible for the pathogenesis of diabetic complications and the development of micro-vascular diseases (Aronson, 2008, Das Evcimen and King, 2007). Retinal microvascular endothelial cell (REC) proliferation and neovascularization at the back of the eyes are hallmarks of proliferative diabetic retinopathy (PDR) (Fong et al., 2004, Moss et al., 1998), and dysfunction of the RECs contributes to the pathogenesis of micro-vascular complications in diabetes. Generally, numerous growth factors such as vascular endothelial growth factor (VEGF) are reported to increase in patients with PDR (Selim et al., 2010), in the retinas of diabetic rats (Jin et al., 2010, Yu et al., 2010) and in high-glucose-induced endothelial cells (Li et al., 2010, Sun et al., 2010). Thus, VEGF is considered to be a key modulator of the vascular alterations in DR pathology. Moreover, VEGF promotes the proliferation of RECs. The excessive secretion of retinal VEGF stimulates neovascularization or angiogenesis in the eyes of diabetic patients, whereas the specific inhibition of VEGF prevents these complications in animal models (Qaum et al., 2001). There are already several reports that show the expression of VEGF in the retinas of STZ-induced diabetic rats, and a number of preclinical and clinical studies have shown a very strong correlation between increased VEGF expression and the development of PDR in response to hyperglycemia or hypoxia (Aiello et al., 1994). However, even under tight glycemic control, the number of DR patients continues to grow, and the therapeutic options are very limited (Ioacara et al., 2009). Therefore, it is necessary to understand the molecular mechanisms underlying the altered expression of VEGF during the pathogenesis of DR and to develop effective measures for treating and preventing RECs proliferation and neovascularization.

MicroRNAs (miRNAs) are a well-recognized group of small (21–25 nucleotides), single stranded, noncoding RNA molecules that bind to the 3′ untranslated region (3′ UTR) of specific target transcript, regulating mRNA activity by inducing their degradation or suppressing their translation (Bartel, 2009, He and Hannon, 2004). Previous studies have shown that miRNAs are involved and play critical roles in a variety of biological processes in the endothelial cells, including their proliferation (Suarez et al., 2007), migration (Sabatel et al., 2011), and survival (Zhang et al., 2009). Recently, miRNAs are shown to be potential mediators of glucose responses (Patella and Rainaldi, 2012). Our previous work using miRNA expression profiling of RECs in normal and diabetic rats indicates that several miRNAs are significantly down-regulated in the RECs of diabetic rats compared with controls, suggesting that miRNAs might be important in the regulation of the pathogenesis of DR (Wu et al., 2012, Kovacs et al., 2011). However, the biological roles of miRNAs in endothelial dysfunction and DR induced by hyperglycemia are still largely unknown and require further investigation.

Insulin-like growth factor-1 (IGF-1) has mitogenic actions in several cells and tissues, including retina (Spoerri et al., 2003). It is reported that IGF-1 possesses a key role in retinal development and pathological states, such as diabetic retinopathy (Hellstrom et al., 2003). In addition, elevated vitreal and serum levels of IGF-1 have been reported to contribute to PDR in patients and also, are known to stimulate endothelial cell proliferation in vitro and in vivo (Payne et al., 2011). Moreover, IGF-1 can act directly as a mitogenic factor on endothelial cells and indirectly induce neovascularization by regulating the expression of potential factors including VEGF (Ruberte et al., 2004). Abundant evidences suggest that the IGF family is a multicomponent network of molecules in regulating pathophysiological growth processes in DR. For example, when RECs exposed to high glucose, IGF-1 binding to insulin growth factor-1 receptor (IGF-1R) activates the phosphatidylinositol 3 kinase (PI3K) or mitogen-activated protein kinase (MAPK) pathway, and then promotes the proliferation (DeBosch et al., 2002, Panjala and Steinle, 2011). IGF-1 participates in the pathogenesis of DR by activating the PI3K/Akt signaling pathway and inducing retinal VEGF expression (Poulaki et al., 2004). Though there is strong experimental and clinical evidence indicating that IGF-1 is associated with the development of DR, the regulated mechanism of IGF-1 in pathogenesis of DR is not well understood and remains controversial.

Based on our previous work of miRNA differential expression in retinal tissues from normal and STZ-induced diabetic rats (Wu et al., 2012), we identified that microRNA-18b (miR-18b) was significantly decreased in DR. To clarify whether miR-18b played a role in DR induced by hyperglycemia, we assessed the effects of gaining and/or losing miR-18b in primary HRECs that cultured in both normal and high glucose mediums. We found that the decrease of miR-18b in high-glucose-induced HRECs promoted cell proliferation and VEGF production. Notably, we demonstrated that miR-18b performed its function by targeting IGF-1, consequently reducing activation of the pathway downstream of IGF-1/IGF1R, suggesting a potential new therapeutic target in the treatment of DR.

Section snippets

HRECs culture and identification

HRECs were respectively isolated from the eyes of 4 donors (Table 1) who had traffic accidents and were treated at either Changhai hospital or Renji hospital as previously described (Chen et al., 2013). HRECs isolated methods were carried out in accordance with the approved guidelines. This study was approved by the Ethics Committee of the Changhai Hospital of the Second Military Medical University and Renji hospital. Retinas from four different donors were respectively harvested, cut into

High glucose promotes the proliferation of HRECs

Primary HRECs were isolated from adult humans as described in Section 2 of this paper. HRECs were respectively identified using morphological and immunofluorescence analyses followed with staining by endothelial markers. Primary HRECs from all four donors exhibited a cobblestone-like morphology and could be passaged without any observable morphological changes (Fig. 1A). Furthermore, cultured HRECs were positive for CD31, von Willebrand factor (vWF) and VE-cadherin, all of which were biological

Discussion

DR is a major complication of both type I and type II diabetes during the later phase and is present in almost every patient who has been diabetic for over 15 years (Ioacara et al., 2009). DR can be divided into two stages, non-proliferative and proliferative, according to its development. Abnormal proliferation of RECs is a characteristic of early proliferative DR. HG is considered to be essential for the development of DR. In this study, cultured HRECs under different high-glucose conditions

Conflict of interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This work was funded by the National Natural Science Foundation of China (30801268, 81371044 to Jinhui Wu), the Natural Science Foundation of Shanghai, China (13ZR1409600 to Lin Liu), the Project in Committee of Science of Shanghai (124119a9500 to Lin Liu), and the Project of National Health and Family Planning Commission of the PRC (PW2013D-1 to Lin Liu).

References (54)

  • A. Abbas et al.

    Role of IGF-1 in glucose regulation and cardiovascular disease

    Expert Rev. Cardiovasc. Ther.

    (2008)
  • L.P. Aiello et al.

    Vascular endothelial growth factor in ocular fluid of patients with diabetic retinopathy and other retinal disorders

    N. Engl. J. Med.

    (1994)
  • M. Al-Latayfeh et al.

    Antiangiogenic therapy for ischemic retinopathies

    Cold Spring Harb. Perspect. Med.

    (2012)
  • D. Aronson

    Hyperglycemia and the pathobiology of diabetic complications

    Adv. Cardiol.

    (2008)
  • Y.C. Chan et al.

    Downregulation of endothelial microRNA-200b supports cutaneous wound angiogenesis by desilencing GATA binding protein 2 and vascular endothelial growth factor receptor 2

    Arterioscler. Thromb. Vasc. Biol.

    (2012)
  • H.S. Chen et al.

    Interaction between glycaemic control and serum insulin-like growth factor 1 on the risk of retinopathy in type 2 diabetes

    Eur. J. Clin. Invest.

    (2012)
  • X. Chen et al.

    KH902 suppresses high glucose-induced migration and sprouting of human retinal endothelial cells by blocking VEGF and PIGF

    Diabetes Obes. Metab.

    (2013)
  • B.J. DeBosch et al.

    Insulin-like growth factor-1 effects on bovine retinal endothelial cell glucose transport: role of MAP kinase

    J. Neurochem.

    (2002)
  • B. Feng et al.

    miR133a regulates cardiomyocyte hypertrophy in diabetes

    Diabetes Metab. Res. Rev.

    (2010)
  • D.S. Fong et al.

    Diabetic retinopathy

    Diabetes Care

    (2004)
  • R. Gao et al.

    Scutellarein inhibits hypoxia- and moderately-high glucose-induced proliferation and VEGF expression in human retinal endothelial cells

    Acta Pharmacol. Sin.

    (2008)
  • M. Guma et al.

    Genetic and pharmacological inhibition of JNK ameliorates hypoxia-induced retinopathy through interference with VEGF expression

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • L. He et al.

    MicroRNAs: small RNAs with a big role in gene regulation

    Nat. Rev. Genet.

    (2004)
  • A. Hellstrom et al.

    Postnatal serum insulin-like growth factor I deficiency is associated with retinopathy of prematurity and other complications of premature birth

    Pediatrics

    (2003)
  • B. Kovacs et al.

    MicroRNAs in early diabetic retinopathy in streptozotocin-induced diabetic rats

    Invest. Ophthalmol. Vis. Sci.

    (2011)
  • A. Krupa et al.

    Loss of MicroRNA-192 promotes fibrogenesis in diabetic nephropathy

    J. Am. Soc. Nephrol.

    (2010)
  • Y. Pan et al.

    Platelet-secreted microRNA-223 promotes endothelial cell apoptosis induced by advanced glycation end products via targeting the insulin-like growth factor 1 receptor

    J. Immunol.

    (2014)
  • Cited by (0)

    View full text