Abstract
AIM
To explore the susceptible association between the insulin-like growth factor-1 receptor (IGF1R) single nucleotide polymorphism (SNP) and age-related cataract (ARC), and investigate the underlying mechanisms in human lens epithelium (HLE) cells.
METHODS
Totally 1190 unrelated participants, comprising 690 ARC patients and 500 healthy individuals in Han Chinese population were recruited and genotyped for target SNP. The χ2-test was used to detect genotypic distribution between the patient and control groups and the logistic regression was performed to adjust the age and gender. Meanwhile, different biological experimental methods, such as cell counting kit 8 (CCK-8) assay, flow cytometry, quantitative real time polymerase chain reaction (Q-PCR) and Western blot, were used to detect cell viability, cell cycle progression and apoptosis in HLE cells or IGF1R knockdown HLE cells.
RESULTS
The rs1546713 in IGF1R gene was identified (P=0.046, OR: 1.606, 95%CI: 1.245-2.071), which shown a significant relevance with ARC risk under the dominant model. The results demonstrated that IGF1R knockdown inhibited cell proliferation by inducing cell cycle arrested at S phase and promoting apoptosis. Mechanistically, the cell cycle blocked at S phase was linked with the alterations of cyclin A, cyclin B, cyclin E and P21. The pro-apoptosis function of IGF1R may related with stimulating the activation of Caspase-3 and altering the expression levels of apoptotic proteins, including Bcl-2, Bax and Caspase-3.
CONCLUSION
This study first report that IGF1R polymorphisms may affect susceptibility to ARCs in Han Chinese population and provide new clues to understand the pathogenic mechanism of ARCs. Notably, IGF1R is likely a potential target for ARC prevention and treatment.
Keywords: insulin-like growth factor-1 receptor, age-related cataract, single nucleotide polymorphism
INTRODUCTION
Cataracts are the leading cause of blindness[1]–[2], resulting in more than 50% of the vision loss worldwide, including 33.4% of sight-disabled people and 18.4% of people with varying degrees of visual impairments[3]–[4]. More than half of elderly people over 65 years old suffer from age-related cataracts (ARCs)—the most common cataract type[5]. Recently, genetic factor, especially single nucleotide polymorphisms (SNPs) becomes a popular topic, which plays a crucial role in the development of ARCs[6]–[7]. Consequently, many genes and SNPs related to ARCs have been identified[8]. Although genetic variation has become the focus of research, the definite pathogeny of ARCs is not completely clear.
The insulin-like growth factor-1 receptor (IGF1R) gene, a member of the tyrosine kinase receptor superfamily[9], is located at 15q26 (MIM:147370). This gene encodes the IGF1R protein, which is composed of two extracellular alpha subunits and two intracellular beta subunits[10]. It regulates numerous downstream signals and is usually involved in tumorigenesis, survival and metastasis by regulating the cell growth and apoptosis of tumour cells[11]–[13], but the effects of IGF1R on cataracts have not been investigated.
In this study, we first found an association between IGF1R SNP and ARCs in ARC patients and explored the impact of IGF1R in human lens epithelium (HLE) cells. These data provide new information for understanding the pathogenic mechanism of ARCs, and notably, IGF1R is likely a potential therapeutic target for ARCs[4].
SUBJECTS AND METHODS
Ethical Approval
This study strictly complied with the Declaration of Helsinki and was approved by the Ethics Committee of the Second Affiliated Hospital, Zhejiang University School of Medicine. Each participant received a full explanation of the purpose and procedures of the research and then signed an informed consent form.
Study Participants
Totally 1190 participants, 690 ARC patients and 500 healthy controls, were recruited in this study. All of them were unrelated Han Chinese and recruited from their visit to the Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine.
All subjects were undergone a full ophthalmologic examination firstly and diagnosed with ARC by the lens opacity classification system II (Locs II)[14]. The ARC patients were divided into four subgroups: cortical cataract (C), posterior subcapsular cataract (PSC), nuclear cataract (N) and mixed cataract (M), based on the range of the lens opacity. Exclusive criteria: patients with diabetes, glaucoma, trauma or other reasons resulting in secondary cataracts. The control subjects were healthy individuals whose lenses were transparent and without other diseases.
Haplotype
The linkage disequilibrium blocks and the haplotype frequencies were tested via the Haploview software. The haplotype around the rs1546713 with D′ values >0.9.
Statistical Analysis
The continuous variables of the subject characteristics were analysed by independent-sample t-test to detect the correlations between groups, and the discontinuous variables were analysed by χ2 test. The χ2 test was performed to compare the genotypic distributions between the case and control groups and to estimate odds ratios (ORs) and 95% confidence intervals (CIs) using version 11.0 SPSS software (USA) and also used to evaluate the Hardy-Weinberg equilibrium (HWE) of SNP using PLINK (v1.07). A logistic regression analysis was executed to adjust for age and sex. To cut down the rate of type I errors, a Bonferroni correction was conducted. In addition, the susceptible relevance between ARCs and SNPs was detected under diverse genetic models: additive, recessive and dominant. The P value <0.05 was considered statistically significant.
Cell Culture and Transfection
The HLE cell line was obtained from the RIKEN Cell Bank (SRA01-04, Tsukuba, Japan) and were maintained in Dulbecco's modified Eagle's medium (DMEM, Corning, NY, USA) with 10% foetal bovine serum (FBS, Biological Industries, Israel) and a 1% penicillin-streptomycin solution (Gibco, Waltham, MA, USA) in a humidified incubator at 37°C. The cells were cultured in six-well plates and added a mixture of 7.5 µL Lipofectamine TM 3000 reagent (Invitrogen, Carlsbad, CA, USA) and 10 µL IGF1R siRNA (100 nmol/L) or NC siRNA until the cell density reached to 30%.
Cell Proliferation Assay
The alteration of cell growth rate was assessed by cell counting kit 8 (CCK-8) assay (Dojindo, Kyushu, Japan). The HLE cells were allowed to adhere to a 96-well plate at a concentration of 1×104 cells/mL overnight. The IGF1R siRNA groups and NC groups were transfected with IGF1R siRNA and NC for 24, 48, and 72h, respectively. Then CCK-8 reagent was added for 2h at 37°C. Finally, the cell proliferation was calculated based on the optical density (OD) value, which was tested by microplate reader at 450 nm.
Apoptosis Assay
The cells were collected and resuspend softly in 1×binding buffer at a density of 1×106 cells/mL. Next, using Annexin V-FITC and PI (BD Pharmingen, San Diego, USA) stained the cells for 15min in the dark at room temperature. Subsequently, the apoptotic cells were tested via a CytoFLEX LX cytometer.
Cell Cycle Analysis
The HLE cells were collected and suspended in 1 mL DNA staining solution and 5 µL permeabilization solution (CCS012, Multi Sciences, Hangzhou, China) and then incubated in the darkroom for 30min. Finally, the samples were analysed by a CytoFLEX LX cytometer.
Quantitative Real-time Polymerase Chain Reaction Analysis
Using 1 mL of Trizol reagent to extracte the total RNA, and transcribing 500 ng of RNA into cDNA. Quantitative real time polymerase chain reaction (Q-PCR) was conducted with a ChamQ SYBR qPCR Master Mix kit (Q311-02, Vazyme, Nanjing, China) and 7500 Fast Real-time PCR System. The sequence of primers is summarizing in Table 1. The relative RNA levels were analysed using the 2−ΔΔCt quantification method after normalization to β-actin.
Table 1. Sequences of forward and reverse primers used in Q-PCR.
| RNA species | Primer pairs |
| IGF1R | Forward: 5′-GAGTGCTGTATGCCTCTGTGAAC-3′ |
| Reverse: 5′-CCTTGGCAACTCCTTCATAGAC-3′ | |
| Bcl-2 | Forward: 5′-GCCACTTACCTGAATGACCACC-3′ |
| Reverse: 5′-AACCAGCGGTTGAAGCGTTCCT-3′ | |
| Bax | Forward: 5′-AGACACCTGAGCTGACCTTGGAG-3′ |
| Reverse: 5′-GTTGAAGTTGCCATCAGCAAACA-3′ | |
| CyclinA | Forward: 5′-TAGCTGCTCCAACAGTAAATCAG-3′ |
| Reverse: 5′-AGGTATGGGTCAGCATCTATCAA-3′ | |
| CyclinB | Forward: 5′-TGCAGCAGGAGCTTTTTGCT-3′ |
| Reverse: 5′-CCAGGTGCTGCATAACTGGAA-3′ | |
| CyclinE | Forward: 5′-ATCAGCACTTTCTTGAGCAACA-3′ |
| Reverse: 5′-TTGTGCCAAGTAAAAGGTCTCC-3′ | |
| P21 | Forward: 5′-CCTGTCACTGTCTTGTACCCT-3′ |
| Reverse: 5′-GCGTTTGGAGTGGTAGAAATCT-3′ |
Q-PCR: Quantitative real time polymerase chain reaction.
Western Blot Assay
The cells were collected and lysed for 30min. The protein was extracted and mixed with 1× SDS sample buffer. Equal quantity of samples were electrophoresed by a 12% SDS-PAGE gel and then transferred onto 0.22-µm PVDF membranes. Used 5% (w/v) non-fat milk in TBST as a blocking agent for two hours and then incubated the membranes with primary antibodies (9665T, anti-Caspase-3, 1:1000; 2872T, anti-Bcl-2, 1:1000; 5023T, anti-Bax, 1:1000 from CST; ab52866, anti-α-Tubulin, 1:1000 from Abcam) at 4°C overnight. Then, with the appropriate secondary anti-bodies for one hour. The blots were detected by ECL chemiluminescence detection kit (Vazyme, Nanjing, China).
Caspase-3 Activity Assay
The HLE cells and IGF1R knockdown HLE cells were lysed and extracted the supernatant. Then, Bradford Protein Assay was used to test the concentrations of sample. The proteins and Ac-DEVD-pNA (2 mmol/L, Beyotime Biotechnology) were mixed at a certain proportion and then incubated for 12h at 37°C. The OD value was detected by the absorbance at 405 nm. Finally, the result was calculated as the OD value/µg protein.
RESULTS
Characteristics of the Participants
This study comprised 690 ARC patients and 500 control subjects. The age or sex between the two groups had no significant difference (P>0.05; Table 2).
Table 2. The characteristics of the participants included in this study.
| Group | n | Gender |
Age (y) |
||
| Male (%) | Female (%) | Mean±SD | Range | ||
| Control | 500 | 58.80 | 41.20 | 64.306±7.586 | 49-92 |
| ARC | 690 | 47.83 | 52.17 | 66.323±9.781 | 43-91 |
| N | 126 | 38.10 | 61.90 | 68.259±9.856 | 45-87 |
| C | 131 | 34.35 | 65.65 | 67.75±8.383 | 43-88 |
| PSC | 73 | 45.21 | 54.79 | 66.484±10.274 | 45-90 |
| M | 360 | 56.11 | 43.89 | 65.170±9.936 | 38-91 |
ARC: Age-related cataract; C: Cortical cataract; N: Nuclear cataract; PSC: Posterior subcapsular cataract; M: Multiple cataract.
Bioinformatics Characteristics of Target SNP
The target SNP in IGF1R were genotyped (Table 3). The target SNP in this research were consistent with HWE.
Table 3. The bioinformatics characteristics of the target SNP.
| SNP | Minor allele | Call rate | MAF | Test for HWE (P) | Control (MAF) | ARC (MAF) |
| rs1546713 | A | 0.9974 | 0.367 | 0.516 | 0.365 | 0.369 |
Relevance Between the Target SNP and Risk of ARCs
The IGF1R target SNP (rs1546713, pa=0.046, OR 1.606, 95%CI 1.245-2.071) showed a notable correlation with general type ARC susceptibility under the dominant model (Table 4).
Table 4. The association between target SNP and ARC risk.
| Cataract type | Logistic regression |
|
| P/pa | OR (95%CI) | |
| rs1546713 | ||
| Dominant | ||
| General | 2.69×10−4/0.046 | 1.606 (1.245, 2.071) |
| C | - | - |
| N | - | - |
| PSC | - | - |
| M | - | - |
| Recessive | ||
| General | - | - |
| C | - | - |
| N | - | - |
| PSC | - | - |
| M | - | - |
| Additive | ||
| General | - | - |
| C | - | - |
| N | - | - |
| PSC | - | - |
| M | - | - |
The Haplotype Around rs1546713
The Haplotype around rs1546713 were inferred by Haploview software to test the linkage disequilibrium (LD) block structure. As shown in Table 5, the LD block located in IGF1R gene.
Table 5. The Haplotype around rs1546713 in a Han Chinese population.
| Haplotype | Frequency |
| CCTAT A CATGAGTAAGAACTCGGTCGCCCAAC | 0.368 |
| TTCCT G TGCATACGGAGGTCTTCCTGCTCAGC | 0.281 |
| TTCAG G TATATACGGAGGTCTGCCTATTTGGT | 0.214 |
| TTCCT G TGTATACGGAGGTCTTCCTGCTCAGC | 0.067 |
Regulatory Effects of IGF1R Knockdown on Cell Proliferation and the Cell Cycle in HLE Cells
IGF1R knockdown HLE cells were constructed by siRNA (transfected for 72h), and the silencing efficiency was confirmed via Q-PCR, with 63% decrease in mRNA expression level of IGF1R (P<0.001, Figure 1A). Next, to explore the effects of IGF1R silence on cell proliferation in HLE cells, the CCK-8 assay was used. As shown in Figure 1B, cell growth was suppressed by IGF1R knockdown compared to NC groups (P<0.05). In addition, to determine the mechanism of the anti-proliferation function of IGF1R knockdown, we detected the influence of IGF1R siRNA on cell cycle progression via flow cytometry analysis. As shown in Figure 1C, a lessened proportion of the G0/G1-phase cells were observed from 81.05%±0.68% to 71.85%±1.24%, and the proportion of G2/M-phase cells was slightly declined from 10.24%±1.85% to 9.78%±1.18% for the NC groups compared to the IGF1R groups. However, a remarkable accumulation of HLE cells in the S phase was noticed from 7.87%±1.04% (NC groups) to 17.28%±0.39% (IGF1R siRNA groups; P<0.05; Figure 1D), which indicated that IGF1R knockdown blocked the cell cycle at the S phase. Moreover, to further confirm the above results, we detected the expression of S phase-related molecules using Q-PCR. After IGF1R knockdown, the cyclin A, cyclin E and P21 mRNA expression level were risen, and the cyclin B mRNA expression level was declined (P<0.05; Figure 1E). These findings demonstrated that the knockdown of IGF1R inhibited cell proliferation by inducing S-phase arrest and altered S-phase-related molecules.
Figure 1. IGF1R-knockdown inhibits the cell proliferation and induced cell cycle arrested in HLE cells.
A: The silence efficiency of IGF1R siRNA; B: The alteration of cell proliferation after treatment for 24, 48, 72h, and a significant difference was observed after 48h; C: The cell cycle analysis in IGF1R knockdown HLE cells; D: Bar diagram of different cell phase in HLE cells; E: The mRNA expression of S-phase related moleculars. All data presented the means±SD from three independent experiments. aP<0.05, bP<0.01, cP<0.001 indicted a significant difference, compared with the NC groups.
Effects of IGF1R on apoptosis in HLE cells
To determine whether IGF1R is associated with ARCs by affecting the apoptosis process, we conducted an Annexin V-FITC/PI staining assay. In comparison with the control, IGF1R siRNA groups had remarkably increased apoptosis, with 9.13% and 22.24% apoptotic cell rates, respectively (P<0.05; Figure 2A-2B). To further verify the authenticity of the experimental data, we separately measured the expression of apoptosis-related mRNAs and proteins through Q-PCR and Western blot assays. As shown in Figure 2C and 2D, the analysis indicated that Caspase-3 and Bax expression levels were distinctly increased in the IGF1R siRNA groups, expect for the decreased expression of the Bcl-2 (P<0.05). The Q-PCR results indicated similar results, that the mRNA level of Bax was significantly increased, but the level of Bcl-2 mRNA was reduced after IGF1R knockdown (P<0.05; Figure 2E). Additionally, as shown in Figure 2F, we also found that Caspase-3 activity increased after treatment with IGF1R siRNA.
Figure 2. IGF1R-knockdown induced apoptosis and altered the apoptosis-related molecule expression and Caspase-3 activities in HLE cells.
A: The apoptotic events after transfected with siRNA; B: The apoptotic proportion of IGF1R knockdown HLE cells; C: The apoptosis-related protein expression level; D: The bar diagram of expression levels of apoptosis-related protein; E: The Bax and Bcl-2 mRNA expression levels; F: The Caspase-3 activities. All data are shown as means±SD from three independent experiments. aP<0.05, bP<0.01 indicated a significant difference, compared with the NC groups.
The above results revealed that IGF1R knockdown promoted apoptosis in HLE cells. Moreover, the proportion of S phase-cells was notably risen, which indicated that the effects of IGF1R knockdown on inducing cell apoptosis were possibly related to the arrested cell cycle progression in the S phase.
DISCUSSION
IGF1R, a type 2 tyrosine kinase transmembrane receptor[15], consists of two α and two β subunits and has two major ligands, IGF1 and IGF2. Once activated, the α subunits induce tyrosine autophosphorylation of the β subunits and then activate and regulate various downstream signalling pathways within cells[16]. IGF1R is expressed in numerous cell types and is crucial for controlling proliferation during growth progression. Homozygous IGF1R-/- mice die at birth and exhibit a serious growth defect, reaching only 45% of the normal size[17]–[18]. A study of zebrafish embryos stressing the importance of IGF1R in the eyes suggested that IGF1R is indispensable for lens development[19]. Other studies shown that the IGF1R expressed in lens epithelial and fiber cells of adult chicken, which in charge of phosphorylating substrate when the hormones binding[20]. Besides, the expression of the IGF1R gene in the human lens varies by age, even in the human embryo stage, and is slightly decreased at 65y and older. IGF1R seems to be age-related, in accordance with the decreased proliferative capacity of HLE cells in elderly persons[21]. These findings may provide new insights into the effects of IGF1R in lens.
As mentioned above, IGF1R is essential for lens development, but no previous study has linked IGF1R polymorphisms with the risk of ARCs. In the current study, we first identified rs1546713 in the IGF1R gene, which is significantly relevant to an altered risk of ARCs under the dominant model in the Han Chinese population. Generally, variants in lens proteins result in rapid and direct protein aggregation and usually lead to cataract formation. Conversely, genetic mutations merely increase susceptibility to environmental hazards and tend to cause ARC through the accelerated accumulation of damage to lens proteins[22]. Thus, the identification of genetic variants for ARC risk is becoming increasingly important. Finding potentially risky genes and their signalling pathways will provide therapeutic targets for the prevention of ARC and improve the efficiency of screening populations with incipient lens opacity; these targets could even help to directly identify susceptible individuals through genetic testing to start therapeutic measures as early as possible[23]. Additionally, to explore its mechanisms, we investigated the impact of IGF1R on cell proliferation, cell cycle and apoptosis in HLE cells, which resulted in new information for understanding the pathogeny of ARC.
In the present study, we found that in IGF1R-knockdown HLE cells, the cell growth rate was remarkably decreased. These findings supported that IGF1R promoted cell proliferation in HLE cells, and Kato et al[24] also reported the pro-proliferation function of IGF1R in prostate cancer cells. Moreover, cell cycle progression is one of the immediate cause of affecting cell proliferation[25]. Therefore, to explore the mechanism of the anti-proliferation effects induced by IGF1R siRNA, we detected the process of HLE cell cycle. As shown in the data, a higher proportion of HLE cells was obviously arrested at the S phase in the IGF1R siRNA groups compared to that in the NC groups. Although our results differ from some studies that the blockage of IGF1R by IGF1R knockdown or the addition of picropodophyllin (an IGF1R-specific inhibitor) resulted in cell cycle arrest at the G2/M phase. However, Sell et al[26] found that IGF1R affected each stage of the cell cycle by testing the distributions between wild-type and IGF1R knockdown mouse embryo fibroblasts. Thus, the different effects of IGF1R on cell cycle progression are likely attributed to different cell types. Furthermore, cell cycle progression is regulated by cyclins[27]. Therefore, we examined S phase-related molecules using Q-PCR. The expression level of cyclin A mRNA, which is essential for S phase initiation and DNA synthesis[28]–[29], were increased. In addition, the high level of cyclin A upon S phase arrest appears to be associated with DNA damage. Several studies suggested that the cyclin A level decreased when the cell cycle arrested at the S phase, but Preya et al[30] noted that α-terthienylmethanol remarkably promoted cyclin A expression when S phase arrest was induced. Similarly, we also found that IGF1R knockdown blocked the cell cycle at the S phase and simultaneously increased cyclin A expression in HLE cells[31]. Cyclin E, responsible for the transition from G0/G1 to S phase, was also increased. Simultaneously, the lower expression of cyclin B mRNA was in response to the fact that HLE cells that could not enter the G2/M phase easily[28],[32]. P21WAF1/CIP1, a CDK inhibitor (CKI), and P21WAF1/CIP-overexpression may induce cell cycle arrest at any phase. A 2.5-fold increase was observed after transfection with IGF1R siRNA. Thus, the above data suggested that IGF1R silencing inhibited cell proliferation by inducing the blockage of the cell cycle at the S phase and affecting S phase-related molecule expression.
The cell cycle and apoptosis are two major mechanisms regulating cell proliferation. Interestingly, when the cell cycle arrests at specific checkpoints, apoptotic events occur[33]–[34]. It has been reported that some anti-cancer drugs act mainly by blocking the cell cycle at the G0/G1, S or G2/M phase to cause apoptosis. There are no data focusing on how IGF1R is related to apoptosis in HLE cells, but apoptosis is a normal physiological phenomenon and is responsible for cataractogenesis. Several pieces of clinical evidence have revealed that the apoptotic HLE cells of ARC patients are remarkably higher than those of healthy people[35]. It is widely accepted that the accumulation of damage from UV, oxidative stress and toxic agents triggers apoptosis in HLE cells[36]. Due to HLE cell death via apoptosis, cell growth will slow, leading to lower cell density and a thinner lens[37]. In addition, the breakdown of the lens epithelial cell barrier, which acts as an umbrella to protect the underlying fibre cells from impairment, will contribute to deficiencies in defence systems against stimuli and interfere with the integrity and transparency of fibre cells, thus resulting in lens opacification. These studies demonstrated that the apoptosis of lens epithelial cells is a common cellular basis for the formation of non-congenital cataracts[37], and there may be a potential therapeutic strategy for ARCs. In the present study, we detected apoptosis via an Annexin V-FITC kit in HLE cells. A significantly higher proportion of apoptotic cells was observed after transfection with IGF1R siRNA compared with that in the NC groups. These results accord with the previous data presented that the activation of IGF1R is able to protect glioblastoma cancer cells from apoptosis[11]. To explore its molecular mechanism, we detected apoptosis-related molecules via Western blot and Q-PCR assays. Caspase-3, which is a key executor of cell dismantling that is essential for the development of apoptotic bodies[38] and positively correlates with apoptotic levels, was distinctly increased in the IGF1R siRNA group. Bax, one of the most important proapoptotic genes, mediates the mitochondrial outer membrane (MOM) permeabilization, resulting in the release of proapoptotic factors[39]–[40], is also upregulated. However, a less Bcl-2 expression, which is an anti-apoptotic gene that directly inhibits pro-apoptotic proteins and blocks upstream pro-apoptotic signals[39], was observed in HLE cells after transfection with IGF1R siRNA. Consistent with the Western blot results, the mRNA expression levels of Bax and Bcl-2 were enhanced and suppressed, respectively. Moreover, Caspase-3 activity was also upregulated. These observations suggested that IGF1R knockdown promoted apoptosis by regulating apoptosis-related molecules and altering Caspase-3 activity.
In conclusion, our study first identified the relationship of rs1546713 in IGF1R with ARCs susceptibility in a Han Chinese population. Additionally, to better understand the underlying mechanism of its effects on ARC occurrence, we found that IGF1R knockdown inhibited HLE cell growth through blocked the progression of cell cycle in the S phase and induced the apoptosis in HLE cells, most likely due to the regulation of cell cycle- and apoptosis-related molecules. These results may provide a new basis for IGF1R transmembrane cell signalling in HLE cells for ARC formation. As a result, IGF1R may be a promising target for the treatment of ARCs.
Acknowledgments
Authors' contributions: Cui YL and Yu XN performed the major experiments and wrote the draft of manuscript; Zhang X analysed the genotyping data; Tang YL and Tang XJ revised the manuscript; Yang H, Ping XY and Wu J participate in the experiments; Yin QC, Zhou JY and Xu XY collected blood samples; Shentu XC contributed to the design of the study and provided reagents and materials.
Foundations: Supported by the National Natural Science Foundation of China (No.81670834; No.81970781; No.81800807; No.81800869); the Natural Science Foundation of Zhejiang Province (No.LY17H090004).
Conflicts of Interest: Cui YL, None; Yu XN, None; Zhang X, None; Tang YL, None; Tang XJ, None; Yang H, None; Ping XY, None; Wu J, None; Yin QC, None; Zhou JY, None; Xu XY, None; Shentu XC, None.
REFERENCES
- 1.Bourne RR, Stevens GA, White RA, Smith JL, Flaxman SR, Price H, Jonas JB, Keeffe J, Leasher J, Naidoo K, Pesudovs K, Resnikoff S, Taylor HR. Causes of vision loss worldwide, 1990–2010: a systematic analysis. Lancet Glob Heal. 2013;1(6):e339–e349. doi: 10.1016/S2214-109X(13)70113-X. [DOI] [PubMed] [Google Scholar]
- 2.Resnikoff S, Pascolini D, Mariotti SP, Pokharel GP. Global magnitude of visual impairment caused by uncorrected refractive errors in 2004. Bull World Health Organ. 2008;86(1):63–70. doi: 10.2471/BLT.07.041210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.He M, Wang W, Huang WY. Variations and trends in health burden of visual impairment due to cataract: a global analysis. Invest Ophthalmol Vis Sci. 2017;58(10):4299–4306. doi: 10.1167/iovs.17-21459. [DOI] [PubMed] [Google Scholar]
- 4.Khairallah M, Kahloun R, Bourne R, Limburg H, Flaxman SR, Jonas JB, Keeffe J, Leasher J, Naidoo K, Pesudovs K, Price H, White RA, Wong TY, Resnikoff S, Taylor HR, Vision Loss Expert Group of the Global Burden of Disease Study Number of people blind or visually impaired by cataract worldwide and in world regions, 1990 to 2010. Invest Ophthalmol Vis Sci. 2015;56(11):6762–6769. doi: 10.1167/iovs.15-17201. [DOI] [PubMed] [Google Scholar]
- 5.Klein BE, Klein R, Linton KL. Prevalence of age-related lens opacities in a population. The Beaver Dam Eye Study. Ophthalmology. 1992;99(4):546–552. doi: 10.1016/s0161-6420(92)31934-7. [DOI] [PubMed] [Google Scholar]
- 6.Hammond CJ, Duncan DD, Snieder H, de Lange M, West SK, Spector TD, Gilbert CE. The heritability of age-related cortical cataract: the twin eye study. Invest Ophthalmol Vis Sci. 2001;42(3):601–605. [PubMed] [Google Scholar]
- 7.Hammond CJ, Snieder H, Spector TD, Gilbert CE. Genetic and environmental factors in age-related nuclear cataracts in monozygotic and dizygotic twins. N Engl J Med. 2000;342(24):1786–1790. doi: 10.1056/NEJM200006153422404. [DOI] [PubMed] [Google Scholar]
- 8.Shiels A, Hejtmancik JF. Genetics of human cataract. Clin Genet. 2013;84(2):120–127. doi: 10.1111/cge.12182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu Q, Guan JZ, Sun Y, Le ZY, Zhang P, Yu D, Liu Y. Insulin-like growth factor 1 receptor-mediated cell survival in hypoxia depends on the promotion of autophagy via suppression of the PI3K/Akt/mTOR signaling pathway. Mol Med Rep. 2017;15(4):2136–2142. doi: 10.3892/mmr.2017.6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lee JH, Kim JY, Kim SY, Choi SI, Kim KC, Cho EW, Kim IG. APBB1 reinforces cancer stem cell and epithelial-to-mesenchymal transition by regulating the IGF1R signaling pathway in non-small-cell lung cancer cells. Biochem Biophys Res Commun. 2017;482(1):35–42. doi: 10.1016/j.bbrc.2016.11.030. [DOI] [PubMed] [Google Scholar]
- 11.Zhang MS, Liu JR, Li MJ, Zhang SH, Lu YM, Liang YQ, Zhao K, Li YF. Insulin-like growth factor 1/insulin-like growth factor 1 receptor signaling protects against cell apoptosis through the PI3K/AKT pathway in glioblastoma cells. Exp Ther Med. 2018;16(2):1477–1482. doi: 10.3892/etm.2018.6336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Long L, Rubin R, Baserga R, Brodt P. Loss of the metastatic phenotype in murine carcinoma cells expressing an antisense RNA to the insulin-like growth factor receptor. Cancer Res. 1995;55(5):1006–1009. [PubMed] [Google Scholar]
- 13.Basu S, Rajakaruna S, De Arcangelis A, Zhang LP, Georges-Labouesse E, Menko AS. Α6 integrin transactivates insulin-like growth factor receptor-1 (IGF-1R) to regulate caspase-3-mediated lens epithelial cell differentiation initiation. J Biol Chem. 2014;289(7):3842–3855. doi: 10.1074/jbc.M113.515254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chylack LT, Jr, Leske MC, McCarthy D, Khu P, Kashiwagi T, Sperduto R. Lens opacities classification system II (LOCS II) Arch Ophthalmol. 1989;107(7):991–997. doi: 10.1001/archopht.1989.01070020053028. [DOI] [PubMed] [Google Scholar]
- 15.Iams WT, Lovly CM. Molecular pathways: clinical applications and future direction of insulin-like growth factor-1 receptor pathway blockade. Clin Cancer Res. 2015;21(19):4270–4277. doi: 10.1158/1078-0432.CCR-14-2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lee J, Hong EM, Jang JA, Park SW, Koh DH, Choi MH, Jang HJ, Kae SH. Simvastatin induces apoptosis and suppresses insulin-like growth factor 1 receptor in bile duct cancer cells. Gut Liver. 2016;10(2):310–317. doi: 10.5009/gnl15195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang S, Wang XX, Wu YJ, Han CS. IGF-1R signaling is essential for the proliferation of cultured mouse spermatogonial stem cells by promoting the G2/M progression of the cell cycle. Stem Cells Dev. 2015;24(4):471–483. doi: 10.1089/scd.2014.0376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Liu JP, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r) Cell. 1993;75(1):59–72. [PubMed] [Google Scholar]
- 19.Coppola D, Ouban A, Gilbert-Barness E. Expression of the insulin-like growth factor receptor 1 during human embryogenesis. Fetal Pediatr Pathol. 2009;28(2):47–54. doi: 10.1080/15513810802679498. [DOI] [PubMed] [Google Scholar]
- 20.Jacobs DB, Ireland M, Pickett T, Maisel H, Grunberger G. Functional characterization of insulin and IGF-I receptors in chicken lens epithelial and fiber cells. Curr Eye Res. 1992;11(12):1137–1145. doi: 10.3109/02713689208999539. [DOI] [PubMed] [Google Scholar]
- 21.Bhuyan DK, Reddy PG, Bhuyan KC. Growth factor receptor gene and protein expressions in the human lens. Mech Ageing Dev. 2000;113(3):205–218. doi: 10.1016/s0047-6374(99)00111-6. [DOI] [PubMed] [Google Scholar]
- 22.Shiels A, Hejtmancik JF. Mutations and mechanisms in congenital and age-related cataracts. Exp Eye Res. 2017;156:95–102. doi: 10.1016/j.exer.2016.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ma XY, Jiao XD, Ma ZW, Hejtmancik JF. Polymorphism rs7278468 is associated with age-related cataract through decreasing transcriptional activity of the CRYAA promoter. Sci Rep. 2016;6:23206. doi: 10.1038/srep23206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kato H, Sekine Y, Furuya Y, Miyazawa Y, Koike H, Suzuki K. Metformin inhibits the proliferation of human prostate cancer PC-3 cells via the downregulation of insulin-like growth factor 1 receptor. Biochem Biophys Res Commun. 2015;461(1):115–121. doi: 10.1016/j.bbrc.2015.03.178. [DOI] [PubMed] [Google Scholar]
- 25.Zang QQ, Zhang L, Gao N, Huang C. Ophiopogonin D inhibits cell proliferation, causes cell cycle arrest at G2/M, and induces apoptosis in human breast carcinoma MCF-7 cells. J Integr Med. 2016;14(1):51–59. doi: 10.1016/S2095-4964(16)60238-8. [DOI] [PubMed] [Google Scholar]
- 26.Sell C, Dumenil G, Deveaud C, Miura M, Coppola D, DeAngelis T, Rubin R, Efstratiadis A, Baserga R. Effect of a null mutation of the insulin-like growth factor I receptor gene on growth and transformation of mouse embryo fibroblasts. Mol Cell Biol. 1994;14(6):3604–3612. doi: 10.1128/mcb.14.6.3604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bendris N, Lemmers B, Blanchard JM. Cell cycle, cytoskeleton dynamics and beyond: the many functions of cyclins and CDK inhibitors. Cell Cycle. 2015;14(12):1786–1798. doi: 10.1080/15384101.2014.998085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Coverley D, Laman, Laskey RA. Distinct roles for cyclins E and A during DNA replication complex assembly and activation. Nat Cell Biol. 2002;4(7):523–528. doi: 10.1038/ncb813. [DOI] [PubMed] [Google Scholar]
- 29.Yam CH, Fung TK, Poon RY. Cyclin A in cell cycle control and cancer. Cell Mol Life Sci. 2002;59(8):1317–1326. doi: 10.1007/s00018-002-8510-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Preya UH, Lee KT, Kim NJ, Lee JY, Jang DS, Choi JH. The natural terthiophene α-terthienylmethanol induces S phase cell cycle arrest of human ovarian cancervia the generation of ROS stress. Chem-Biol Interact. 2017;272:72–79. doi: 10.1016/j.cbi.2017.05.011. [DOI] [PubMed] [Google Scholar]
- 31.Itoh T, Ono A, Kawaguchi K, Teraoka S, Harada M, Sumi K, Ando M, Tsukamasa Y, Ninomiya M, Koketsu M, Hashizume T. Phytol isolated from watermelon (Citrullus lanatus) sprouts induces cell death in human T-lymphoid cell line Jurkat cells via S-phase cell cycle arrest. Food Chem Toxicol. 2018;115:425–435. doi: 10.1016/j.fct.2018.03.033. [DOI] [PubMed] [Google Scholar]
- 32.Yu JH, Zhang LY, Ren PY, Zhong T, Li ZL, Wang ZY, Li JL, Liu X, Zhao K, Zhang WY, Yu XF. Enterovirus 71 mediates cell cycle arrest in S phase through non-structural protein 3D. Cell Cycle. 2015;14(3):425–436. doi: 10.4161/15384101.2014.980631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Li Y, Yu HY, Han FF, Wang MM, Luo Y, Guo XJ. Biochanin A induces S phase arrest and apoptosis in lung cancer cells. Biomed Res Int. 2018;2018:3545376. doi: 10.1155/2018/3545376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Orren DK, Petersen LN, Bohr VA. Persistent DNA damage inhibits S-phase and G2 progression, and results in apoptosis. Mol Biol Cell. 1997;8(6):1129–1142. doi: 10.1091/mbc.8.6.1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhou D, Yin D, Xiao F, Hao J. Expressions of senescence-associated β-galactosidase and senescence marker protein-30 are associated with lens epithelial cell apoptosis. Med Sci Monit. 2015;21:3728–3735. doi: 10.12659/MSM.895596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Galichanin K. Exposure to subthreshold dose of UVR-B induces apoptosis in the lens epithelial cells and does not in the lens cortical fibre cells. Acta Ophthalmol. 2017;95(8):834–838. doi: 10.1111/aos.13370. [DOI] [PubMed] [Google Scholar]
- 37.Li WC, Kuszak JR, Dunn K, Wang RR, Ma W, Wang GM, Spector A, Leib M, Cotliar AM, Weiss M. Lens epithelial cell apoptosis appears to be a common cellular basis for non-congenital cataract development in humans and animals. J Cell Biol. 1995;130(1):169–181. doi: 10.1083/jcb.130.1.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6(2):99–104. doi: 10.1038/sj.cdd.4400476. [DOI] [PubMed] [Google Scholar]
- 39.Siddiqui WA, Ahad A, Ahsan H. The mystery of BCL2 family: Bcl-2 proteins and apoptosis: an update. Arch Toxicol. 2015;89(3):289–317. doi: 10.1007/s00204-014-1448-7. [DOI] [PubMed] [Google Scholar]
- 40.Peña-Blanco A, García-Sáez AJ. Bax, Bak and beyond - mitochondrial performance in apoptosis. FEBS J. 2018;285(3):416–431. doi: 10.1111/febs.14186. [DOI] [PubMed] [Google Scholar]


