Diabetic macular edema (DME) remains a leading cause of vision loss worldwide. DME is commonly treated with intravitreal injections of vascular endothelial growth factor (VEGF)–neutralizing antibodies. VEGF inhibitors (anti-VEGFs) are effective, but not all patients fully respond to them. Given the potential side effects, inconvenience, and high cost of anti-VEGFs, identifying who may not respond appropriately to them and why is essential. Herein we determine first the response to anti-VEGFs, using spectral-domain optical coherence tomography scans obtained from a cohort of patients with DME throughout the 1st year of treatment. We found that fluid fully cleared at some time during the 1st year in 28% of eyes (“full responders”); fluid cleared only partly in 66% (“partial responders”); and fluid remained unchanged in 6% (“nonresponders”). To understand this differential response, we generated induced pluripotent stem cells (iPSCs) from full responders and nonresponders, from subjects with diabetes but no DME, and from age-matched volunteers without diabetes. We differentiated these iPSCs into endothelial cells (iPSC-ECs). Monolayers of iPSC-ECs derived from patients with diabetes showed a marked and prolonged increase in permeability upon exposure to VEGF; the response was significantly exaggerated in iPSC-ECs from nonresponders. Moreover, phosphorylation of key cellular proteins in response to VEGF, including VEGFR2, and gene expression profiles, such as that of neuronal pentraxin 2, differed between full responders and nonresponders. In this study, iPSCs were used in order to predict patients’ responses to anti-VEGFs and to identify key mechanisms that underpin the differential outcomes observed in the clinic. This approach identified NPTX2 as playing a significant role in patient-linked responses and as having potential as a new therapeutic target for DME.

Vascular endothelial growth factor (VEGF) inhibitors (anti-VEGFs) are used to treat diabetic macular edema (DME). Anti-VEGFs are given as an ocular injection and are required frequently and over the long-term. The work presented herein demonstrates that in only approximately a quarter of eyes does DME fully resolve during the 1st year of treatment with anti-VEGFs; many eyes do not respond adequately to this therapy. It shows also that in vitro readouts of induced pluripotent stem cell–derived endothelial cell (iPSC-EC) responses can be used to identify a patient’s response to anti-VEGFs; such identification has the potential to improve clinical decision-making about the use of alternative therapies for those who are unlikely to respond to anti-VEGFs. Furthermore, induced pluripotent stem cell (iPSC) technology may enable targeted drug development for DME that is not predominantly driven by VEGF. DME remains a leading cause of visual loss in people with diabetes (1). It results from the breakdown of the inner and outer blood-retinal barriers (BRBs), with subsequent leakage of blood, fluid, and lipids, and infiltration of inflammatory cells into the macula, which alters retinal architecture and function (2). Although the exact mechanisms that lead to DME are not completely understood, evidence suggests that VEGF plays a main role, often in unison with an array of proinflammatory cytokines (3). The amount of VEGF is elevated in the vitreous of patients with DME (4), and experimental studies have demonstrated that VEGF induces vascular leakage (by disrupting the integrity of endothelial cell [EC] junctional complexes) and a loss of barrier function (5).

Since 2010, numerous randomized controlled trials (RCTs) have demonstrated the benefit of VEGF-neutralizing antibodies to improve vision and reduce macular fluid in people with DME (610). Despite the proven efficacy of anti-VEGFs, however, it is now evident that not all patients with DME respond to them in a similar manner. To date, no consensus exists about a definition of “responder” and “nonresponder” with regard to anti-VEGFs (1114), nor is there a robust predictor for who will and will not respond to this therapy. Anti-VEGFs are given as intravitreal injections, and in most patients they need to be administered monthly throughout the 1st year of treatment and at frequent intervals thereafter. Most patients with DME will require long-term treatment in order to maintain the benefits of the anti-VEGF: ∼50% of patients still require treatment 5 years after its initiation (15). An ability to predict a patient’s responsiveness to this therapy would be of great benefit, avoiding unnecessary risks and the inconvenience and distress caused by repeated injections, and allowing more timely intervention with alternative therapies (16). Such a personalized approach to anti-VEGF administration would avoid the loss of vision that may occur as a result of selecting or continuing an ineffective treatment, an unnecessary workload to health professionals, and costs for health services.

In this study, we evaluated the response to anti-VEGFs among a large cohort of patients with DME treated with anti-VEGFs. In only ∼28% of patients did the edema clear fully at any time during the 1st year of anti-VEGF treatment. Also, having previously developed a unique approach to generate iPSCs and differentiated endothelium from small volumes of blood (17), we designed a conjoined clinical and experimental study using iPSC microvascular endothelium derived from patients with diabetic retinopathy (DR) but without DME and from patients with DME who are classified as “full responders” or “nonresponders” to anti-VEGFs. Their iPSC-ECs were assessed for VEGF-induced permeability responses, phosphorylation pathways, and gene expression profiles. The clear differences between full responders and nonresponders may reflect a distinct pathophysiology in ECs from the patient groups and could not only offer useful clinical information but also provide scope for the development of new drugs.

Evaluating the Response to Anti-VEGF Treatment in Patients With DME

Electronic records were retrospectively reviewed for all consecutive patients with newly diagnosed DME who had been examined at the Belfast Health and Social Care Trust (BHSCT). This study was part of an approved audit (audit approval number 5170) for which full ethical review was waived. A patient cohort was constructed. Eligibility criteria for this cohort included 1) receipt of the first dose of anti-VEGF between 1 March and 31 December 2014; 2) treatment exclusively with ranibizumab, as this was the first anti-VEGF agent approved for the treatment of DME in the U.K., and most patients at the BHSCT would have received this drug during the study period; 3) receipt of a minimum loading dose (injections over three consecutive months); and 4) a minimum follow-up of 12 months, with regular visits during that time (every 4–6 weeks).

Spectral-domain optical coherence tomography (SD-OCT) scans obtained throughout the whole 1st year of treatment were reviewed and graded by an expert clinician (N.L.) who was masked to clinical findings. The expert classified patients as “full responders” when the macula fully dried at any point during the 1st year or treatment (although the presence of small, isolated, or sparse cysts was allowed within this definition); as “partial responders” when the amount of fluid present at the macula was reduced but the fluid never fully dried during the 1st year of treatment; or as “nonresponders” when macular fluid remained unchanged upon qualitative evaluation of SD-OCT scans throughout the 1st year of treatment and when the central retinal thickness (CRT) changed ≤15% from the baseline thickness.

Differences were investigated between the aforementioned groups in demographics (age, sex), type of diabetes (type 1 vs. type 2), best-corrected visual acuity, number of injections received, and central retinal subfield thickness measured from SD-OCT scans. The χ2 test was used in order to compare proportions (sex and type of diabetes) among anatomic response categories. Linear regression was fit by using generalized linear models, and SEs were adjusted for within-patient correlation by using a “sandwich” (Huber-White) variance estimator.

Predicting a Response to Anti-VEGF Treatment in Patients With DME by Using iPSC-ECs In Vitro and Elucidating the Molecular Mechanisms Involved

This prospective study was approved by the Office for Research Ethics Committees Northern Ireland under the title of “Stratifying Treatment for Diabetic Macular Oedema Using Induced Pluripotent Stem (iPS) Cells” (STREAMLINE) (reference 14/NI/1109; Integrated Research Application System project identifier 162254). Patients with DR were recruited from ophthalmic clinics at the BHSCT. Volunteers without diabetes were recruited as controls from among individuals accompanying patients to the BHSCT clinic and from Queen’s University Belfast.

Inclusion and Exclusion Criteria

Patients with diabetes and mild nonproliferative DR with no DME (n = 6), those with DME who were classified either as “full responders” (n = 6) or as “nonresponders” (n = 6) to anti-VEGF therapy (ranibizumab), and age-matched volunteers without diabetes (n = 6) were recruited into the study.

Information about the study was provided verbally and in writing to all potentially eligible participants. Informed consent was obtained from those who were willing and who consented to take part, before beginning the study procedures. Patients unable to provide informed consent for the study were excluded. The study adhered to the principles of ethical medical research as detailed in the Declaration of Helsinki.

Blood Sampling and Masking

On the day blood samples were collected, a detailed medical and ocular history was obtained and blood pressure was measured. A 20-mL sample of peripheral blood was obtained from all participants; the samples were collected by venepuncture in VACUETTE 4-mL K3 EDTA-coated tubes (item 454021; Greiner Bio-One International GmbH). Samples were processed in the laboratory by scientists masked to clinical findings and the origin of the samples (i.e., the scientists did not know from which donor group the samples came).

Mononuclear Cell Isolation and Culture

Mononuclear cells (MNCs) were isolated as previously described (17). Briefly, blood was separated by gradient by layering it on Histopaque solution (item 10771; Sigma-Aldrich) at a 1:1 ratio and then centrifuging it at 550g with the break off and at room temperature for 30 min. The MNCs’ buffy coat was collected with a soft plastic pipette and placed in a 15-mL tube, which was topped off with PBS and centrifuged at 300g at 4°C for 10 min. After three washes with PBS, the cells were resuspended in 1 mL of MNC medium, as previously described (17), and plated in MNC medium at a density of 4 million cells per milliliter. The cells were expanded for 7 days and then used for reprogramming.

Generation of Patient-Specific iPSCs

iPSCs were generated from patients’ blood MNCs, as previously described (17). Up to 2 million MNCs were transfected with 5 μg of plasmid per million cells (a 5:1 ratio of pEB-C5 and pEB-Tg) by using a Human CD34+ Cell Nucleofector Kit (catalog no. VPA-1003; Lonza) and an Amaxa nucleofector (program T-016), according to the manufacturer’s protocol. pEB-C5 and pEB-Tg plasmids were a gift from Linzhao Cheng (plasmids 28213 and 28220; Addgene). On day 2 after transfection, the cells were seeded onto inactivated feeders (mouse embryonic fibroblasts from ATCC) in reprogramming medium (KnockOut DMEM/F-12 with MEM Non-Essential Amino Acids Solution and GlutaMAX [Thermo Fisher Scientific] supplemented with KnockOut Serum Replacement [20%] [Thermo Fisher Scientific], 0.1 mmol/L 2-mercaptoethanol [Thermo Fisher Scientific], and 10 ng/mL MACS human recombinant fibroblast growth factor 2 [Miltenyi Biotec]).

Reprogramming medium was changed every day and colonies began to appear from day 9. Colonies were individually expanded for the establishment of iPSC lines. For the maintenance of iPSC lines, reprogramming medium was changed every day and the iPSC colonies were subcultured once per week at a 1:6 ratio.

Differentiation of iPSCs into iPSC-ECs

iPSCs were cultured under feeder-free conditions and seeded on Matrigel Growth Factor Reduced Basement Membrane Matrix (catalog no. 354230; Corning Life Sciences) in StemMACS iPS-Brew XF (130-104-368; Miltenyi Biotec). The next day, the medium was replaced with Neurobasal Medium (catalog no. 21103049; Thermo Fisher Scientific) with N-2 Supplement (catalog no. 17502001; Thermo Fisher Scientific) and B-27 Supplement (catalog no. 17504044; Thermo Fisher Scientific) supplemented with 25 ng/mL BMP4 Recombinant Human Protein (catalog no. PHC9531; Thermo Fisher Scientific) and 8 μmol/L CHIR99021 (SML1046; Sigma-Aldrich). The medium was replaced 72 h later with StemPro-34 SFM (catalog no. 10639011; Thermo Fisher Scientific) supplemented with 200 ng/mL VEGF Recombinant Human Protein (catalog no. PHC9391; Thermo Fisher Scientific) and 2 μmol/L forskolin (F6886; Sigma-Aldrich). This step was repeated after 24 h.

On day 6 of differentiation, magnetic activated cell sorting (MACS) of cells expressing the mature EC marker CD144 was performed by using anti–human CD144 (VE-Cadherin) MicroBeads (130-097-857; Miltenyi Biotec). The cells were seeded on plates coated with mouse collagen IV in EGM-2 10% FBS medium supplemented with 50 ng/mL VEGF and 10 μmol/L LY364947. Medium was replaced every other day and the cells were used for further analysis or were cryopreserved once they were confluent.

RNA Extraction, RT-PCR, and Quantitative RT-PCR

Relative mRNA expression of specific cell markers (and other genes of interest) was determined by using RNA extraction, RT-PCR, and quantitative RT-PCR, as previously described (18). Primers for CD235, CD71, OCT4, LIN28, NANOG, CD144, CD31, VEGFR2, and NPTX2 are listed in Supplementary Table 2. GAPDH, ATP5B, ACTB, and UBC were used as reference genes (all from Primer Design Human geNorm Kit [catalog code ge-SY-12; Integrated Sciences]). Sequences were not provided by the manufacturer.

Western Blotting

Proteins of specific cell markers were detected by using Western blotting, as previously described (18). To ensure valid comparison between samples, cells were harvested at the same time point, confluence (70–80% for iPSCs, 80–100% for iPSC-ECs), or both. For the detection of phosphorylated proteins, Tris-buffered saline with Tween was used instead of PBS with Tween, and 5% BSA Tris-buffered saline with Tween was used as a blocking solution instead of 5% milk in PBS with Tween. Antibodies against TRA-1-60 (ab16288), OCT4 (ab19857), LIN28 (ab46020), NPTX2 (ab69858) (all from Abcam), CD144 (STJ96234; St John’s Laboratories), and Phospho-VEGFR2 (PA5-12598) and Phospho-eNOS (PA5-35879) (both from Thermo Fisher Scientific) were used. Antibodies against actin β (MAB8929; R&D Systems) or GAPDH (ab125247; Abcam) were used as the loading control. Western blotting was quantified by using ImageJ software, and samples were normalized to the loading control (actin β or GAPDH).

Immunofluorescence Staining

Immunofluorescence staining procedures have been detailed by Vilà-González et al. (17). Live staining of iPSCs was performed by using CDy1 Dye (14001; Active Motif), as indicated in the manufacturer’s instructions.

Migration Assay

Migration capacity was studied through a wound healing or scratch assay. iPSC-ECs were seeded in wells of a 24-well plate at a density of 50,000 iPSC-ECs per well. Once the cells were confluent, the monolayer was scratched from one side of the well to the other by using a 200-μL pipette tip. Pictures of the wound were taken at the 0-, 4-, 8-, 12-, and 24-h time points. The migration capacity of the cells was calculated by measuring the change from baseline, in which the area of the wound (0 h after the scratch) minus the area of the wound at the various time points (4, 8, 12, and 24 h) equals the area the cells had covered during that time.

Proliferation Assay

Proliferation of iPSC-ECs was assessed by using a CyQUANT NF Cell Proliferation Assay (C35006; Thermo Fisher Scientific), according to the manufacturer’s instructions. iPSC-ECs were seeded in a 96-well plate at a density of 5,000 cells per well and were allowed to adhere for at least 4 h. Cells were seeded in quadruplicate for each cell line and time point (8, 24, and 48 h). The values at the various time points were normalized to the values obtained at 8 h for each experiment.

Permeability Assessment

Permeability response to VEGF in iPSC-ECs from different patients/donors was measured using xCELLigence RTCA DP (Real-Time Cell Analyzer–Dual Purpose; ACEA Biosciences, Inc.), which measures cell impedance in real time. A total of 20,000 cells in 200 μL medium (the medium used to maintain iPSC-ECs EGM-2 10% FBS media supplemented with 50 ng/mL VEGF and 10 mmol/L LY364947) was seeded in each well of a 16-well polyethylene terephthalate plate that had been precoated with mouse collagen IV. Four replicates were seeded for each condition. The cells reached confluence overnight. After seeding (24 h), the cells were starved for 2 h by replacing 150 μL of medium with endothelial basal medium plus HEPES at 25 mmol/L (15630106; Thermo Fisher Scientific). Next, 100 μL of the medium was replaced with starvation medium with 200, 100, or 50 ng/mL VEGF (for a final concentration of 100, 50, or 25 ng/mL). BSA (0.1%) in PBS was used as the vehicle control. Data were analyzed by using RTCA Data Analysis Software and Prism 5 software.

RNA Sequencing

Quality control and RNA sequencing were analyzed at the Queen’s University Genomics Core Technology Unit. In brief, the cells were briefly washed with PBS and harvested by using QIAzol lysis buffer. Samples and the library were prepared as previously described (17). Libraries were sequenced on a NextSeq sequencing system (Illumina), and paired-end reads were mapped to the human reference genome (hg38)—allowing up to 2 mismatches and up to 10 hits per read—by using the CLC Genomics Workbench version 10.0.1 (https://www.qiagenbioinformatics.com). Reference sequences were annotated with genes and transcripts. Reference content was mapped to gene regions only, and the raw read counts were generated by HTSeq-Counts. To identify differentially expressed genes (DEGs) between RNA sequences of samples from responders and nonresponders, a differential expression analysis was performed by using DESeq2 in the R software package, followed by standard visualization by using EnhancedVolcano and Pheatmap in R. Comparative gene expression data were filtered according to the absolute log2 fold change greater than +1 and an adjusted P value (q value) threshold of <0.05.

Lentiviral Particle Transduction and VEGF Treatment

Lentiviral particles were produced by using MISSION shNT (sc-62166; Santa Cruz) or shNPTX2 plasmids (MISSION shNPTX2 (SHCLNG-NM_002523, TRCN0000373047; Sigma-Aldrich) according to a previously described protocol (1820). The efficiency of the infection was 70–80%. Two hours before harvest, the cells were depleted of serum; they were treated with 50 ng/mL VEGF 5 and 20 min before harvest. Vehicle control cells (0.1% BSA in PBS), harvested at exactly 72 h, served as the control (0 min time point).

FITC-Dextran Treatment

iPSC-ECs from nonresponders, which had been seeded in transwell plates, were treated with 1 mg/mL FITC-dextran 4 (46944; Sigma-Aldrich) between 3 and 24 h after transduction with shNPTX2 or shNT. Fluorescence was determined at 520-nm emission and 485-nm excitation, and relative percentage diffusion was calculated.

Statistical Analysis

Data presented in the graphs were analyzed by using GraphPad Prism 5 software with a two-tailed Student t test for two groups or with pairwise comparisons or ANOVA; values are expressed as the mean ± SD. A P value <0.05, <0.01, or <0.001 was considered significant, as indicated.

Data and Resource Availability

The data sets and the patient-specific IPSC lines generated and analyzed during this study are available from the corresponding authors upon reasonable request.

Evaluating the Response to Anti-VEGF Treatment in Patients With DME

A total of 140 eyes from 100 patients with DME were treated with ranibizumab; these eyes/patients formed the study cohort. Color fundus photographs of the right eye were obtained from an individual with no retinal disease (Fig. 1A, left) and from a patient with DME (Fig. 1A, right); hard exudation can be seen involving the macula and extending to the fovea (Fig. 1A, right, black arrow). A total of 172 eyes were treated during the study period; 32 were excluded because they did not meet the eligibility criteria for the study (<12 months of follow-up or use of other intravitreal therapy [i.e., aflibercept or steroids]).

Figure 1

A: Color fundus photographs of the right eye obtained from an individual with no retinal disease (left) and from a patient with DME (right). Hard exudation is seen involving the macula and extending to the fovea (black arrow). B: SD-OCT scans obtained at baseline (top left) and at a time during the 1st year when the retina dried (top right) in a full responder; and at baseline (bottom left) and at the end of the year following repeated treatments (bottom right) in a nonresponder. Intraretinal (top left, white arrow) and subretinal fluid (top left, white arrowhead) are observed at presentation in the full responder; the fluid completely resolved after treatment, and the patient recovered a near-normal macular structure (top panels). Intraretinal fluid (bottom left, white arrow) is seen in a nonresponder; the fluid level changed only minimally throughout the 1st year of treatment (bottom panels).

Figure 1

A: Color fundus photographs of the right eye obtained from an individual with no retinal disease (left) and from a patient with DME (right). Hard exudation is seen involving the macula and extending to the fovea (black arrow). B: SD-OCT scans obtained at baseline (top left) and at a time during the 1st year when the retina dried (top right) in a full responder; and at baseline (bottom left) and at the end of the year following repeated treatments (bottom right) in a nonresponder. Intraretinal (top left, white arrow) and subretinal fluid (top left, white arrowhead) are observed at presentation in the full responder; the fluid completely resolved after treatment, and the patient recovered a near-normal macular structure (top panels). Intraretinal fluid (bottom left, white arrow) is seen in a nonresponder; the fluid level changed only minimally throughout the 1st year of treatment (bottom panels).

Close modal

The mean best-corrected visual acuity was 59.7 Early Treatment Diabetic Retinopathy Study letters (SD, 15.4 letters) at baseline and improved to 67.3 Early Treatment Diabetic Retinopathy Study letters (SD, 14.4 letters) at the 12-month follow-up, after a mean of 7.5 ranibizumab injections (SD, 2.04 injections). The anti-VEGF response observed is summarized in Supplementary Table 3, and the demographic and clinical characteristics of patients (eyes) included in the study are summarized in Supplementary Table 4. Examples of retinal disease in a full responder and in a nonresponder are shown in Fig. 1B. In full responders the macula dried after a median of 7 months (range, 1–12 months).

No differences were found among groups for any of the clinical parameters evaluated other than, as expected, central retinal subfield thickness and the number of intravitreal anti-VEGF injections received. Partial responders and nonresponders received more injections than did full responders. In 39 patients in the cohort, both eyes were eligible and thus were included in the study. Among these patients, concordance of the response between eyes (i.e., right eyes were in the same category of anti-VEGF response as left eyes) was observed in 28 (72%).

Predicting a Response to Anti-VEGF Treatment in Patients With DME By Using iPSC-ECs In Vitro

Demographics and characteristics of participants are detailed in Supplementary Table 1. iPSCs generated from donors with and from those without diabetes showed identical reprogramming and differentiating capacities. These iPSC lines were initially generated from three subjects from each cohort (patients with DR and no DME; patients with DR and DME that responded fully to anti-VEGF treatment [“full responders,” hereafter referred to as “responders”]; those who did not respond to this therapy [“nonresponders”]; and age-matched healthy volunteers). For each group, one iPSC line was selected, expanded, and characterized (Fig. 2) before being differentiated into ECs (iPSC-ECs) (Figs. 3 and 4). Established iPSC colonies were examined for pluripotency by using quantitative RT-PCR (Fig. 2A and B) and immunoblotting (Fig. 2C), which confirmed the high expression of pluripotency markers and the low expression of MNC markers at both the RNA and the protein level. iPSC colonies also stained positive for CDy1, Oct4, Lin28, and TRA-1-60 (Fig. 2D). Overall, iPSCs from the different groups were comparable in terms of expression of pluripotency markers and morphology, indicating the reprogrammable nature of each cohort’s samples.

Figure 2

Generation and characterization of iPSCs from donors without diabetes (IPS nondiabetic), from patients with diabetes and mild DR (IPS mild DR), and from patients with DME (IPS DME) (both responders [R] and nonresponders [NR]). A: Quantitative RT-PCR analysis shows that erythroid markers are no longer present in reprogrammed cells. B: Similar mRNA levels of pluripotent markers were found in all groups. Data are means ± SD (n = 3). *P < 0.05, **P < 0.01. C: Pluripotent markers are also expressed at the protein level in all groups. D: Immunofluorescence staining confirms the characterization of iPSCs in all groups. Scale bars represent 50 μm (white), 100 μm (yellow), or 200 μm (orange).

Figure 2

Generation and characterization of iPSCs from donors without diabetes (IPS nondiabetic), from patients with diabetes and mild DR (IPS mild DR), and from patients with DME (IPS DME) (both responders [R] and nonresponders [NR]). A: Quantitative RT-PCR analysis shows that erythroid markers are no longer present in reprogrammed cells. B: Similar mRNA levels of pluripotent markers were found in all groups. Data are means ± SD (n = 3). *P < 0.05, **P < 0.01. C: Pluripotent markers are also expressed at the protein level in all groups. D: Immunofluorescence staining confirms the characterization of iPSCs in all groups. Scale bars represent 50 μm (white), 100 μm (yellow), or 200 μm (orange).

Close modal
Figure 3

Differentiation and characterization of iPSC-ECs that were successfully generated from patients with diabetes and from donors without diabetes. A and B: Quantitative RT-PCR analysis shows that iPSC-ECs from both groups switched off the expression of pluripotent markers such as OCT4, LIN28, and NANOG (A) and they highly expressed EC-specific markers such as CD144, CD31, and VEGFR2 (B). Data are means ± SD (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001). C: These results were confirmed at the protein level by Western blotting. D: Immunofluorescence staining of endothelial markers confirms these results and demonstrates that iPSC-ECs from both groups were able to form endothelial and adherent junctions. Scale bars = 50 μm. iPS, induced pluripotent stem cells.

Figure 3

Differentiation and characterization of iPSC-ECs that were successfully generated from patients with diabetes and from donors without diabetes. A and B: Quantitative RT-PCR analysis shows that iPSC-ECs from both groups switched off the expression of pluripotent markers such as OCT4, LIN28, and NANOG (A) and they highly expressed EC-specific markers such as CD144, CD31, and VEGFR2 (B). Data are means ± SD (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001). C: These results were confirmed at the protein level by Western blotting. D: Immunofluorescence staining of endothelial markers confirms these results and demonstrates that iPSC-ECs from both groups were able to form endothelial and adherent junctions. Scale bars = 50 μm. iPS, induced pluripotent stem cells.

Close modal
Figure 4

iPSC-ECs were successfully generated from patients with DME (responders [R] and nonresponders [NR]) in a similar way as iPSC-ECs from patients with diabetes and from donors without diabetes. A and B: Quantitative RT-PCR analysis shows that iPSC-ECs from both groups stopped expressing pluripotent markers such as OCT4, LIN28, and NANOG (A) and highly expressed EC-specific markers such as CD144, CD31, and VEGFR2 (B). Data are means ± SD (n = 3). **P < 0.01, ***P < 0.001, ****P < 0.0001. C: These results were confirmed at the protein level by Western blotting. D: Immunofluorescence staining of endothelial markers confirms that iPSC-ECs from patients with DME (both responders and nonresponders) were able to form endothelial and adherent junctions. Scale bars = 50 μm.

Figure 4

iPSC-ECs were successfully generated from patients with DME (responders [R] and nonresponders [NR]) in a similar way as iPSC-ECs from patients with diabetes and from donors without diabetes. A and B: Quantitative RT-PCR analysis shows that iPSC-ECs from both groups stopped expressing pluripotent markers such as OCT4, LIN28, and NANOG (A) and highly expressed EC-specific markers such as CD144, CD31, and VEGFR2 (B). Data are means ± SD (n = 3). **P < 0.01, ***P < 0.001, ****P < 0.0001. C: These results were confirmed at the protein level by Western blotting. D: Immunofluorescence staining of endothelial markers confirms that iPSC-ECs from patients with DME (both responders and nonresponders) were able to form endothelial and adherent junctions. Scale bars = 50 μm.

Close modal

Central to this study was the comparison of patients with DME; therefore, in order to avoid misidentification and to rule out possible cross-contamination of cell lines, short tandem repeat profiling of the iPSC lines was undertaken (Supplementary Table 5). When two cell lines shared less than 56% of the analyzed alleles, they could be considered to come from different individuals; the selected iPSC lines met these criteria (Supplementary Tables 5 and 6), confirming that they came from different donors. Moreover, none of the iPSCs matched any of the lines recorded by the European Collection of Authenticated Cell Cultures. This fact provided important assurance that the iPSCs used in this study were unique and patient-specific.

Differentiation of iPSCs Into iPSC-ECs From Volunteers Without Diabetes and Donors With Diabetes Produces Phenotypically Comparable Cells

Upon iPSC differentiation to iPSC-ECs, flow cytometry demonstrated that before CD144 MACS selection, up to 57.2% of iPSC-ECs were CD144+ (Supplementary Fig. 1); up to 67.2% of cells were CD144+ in the samples from patients with DME (Supplementary Fig. 2). iPSC-ECs from patients with diabetes and volunteers without diabetes displayed a loss of expression of pluripotent markers such as OCT4, LIN28, and NANOG and upregulation of EC markers such as CD144, CD31, and VEGFR2 at the mRNA level (Figs. 3A and B and 4A and B), confirming differentiation toward the vascular lineage. Further characterization by immunoblotting (Figs. 3C and 4C) also confirmed that cells switched off expression of the pluripotency marker TRA-1-60 but highly expressed EC-specific markers such as CD144. There was no difference in EC gene expression and immunophenotype among iPSC-EC groups (Figs. 3 and 4), nor was there a difference, based on immunofluorescent staining, on adherens and tight junction integrity or VEGFR2 expression (Figs. 3D and 4D). These results clearly demonstrate that iPSC-ECs were successfully generated and expanded from all groups (donors without diabetes and patients with diabetes either with or without DME).

iPSC-ECs From Volunteers Without Diabetes and Donors With Diabetes, Including Responders and Nonresponders With DME, Have Similar Phenotypical Characteristics but Different Permeability Responses

iPSC-ECs from donors without diabetes, patients with mild DR, and responders and nonresponders with DME displayed similar functional capacities. The migration capacity of iPSC-ECs from those without and those with diabetes, or from responders and nonresponders, was not significantly different when assessed by using a wound healing assay or scratch assay (Fig. 5A, B, D, and E). In a similar manner, no significant difference in their proliferation capacity was found (Fig. 5C and F). One of the main elements of DME is the excessive permeability of the normally “tight” retinal microvasculature. Therefore, EC permeability was tested in this study after stimulation with VEGF. Significant differences in VEGF-induced cell permeability were identified when cells from patients with diabetes were compared with cells from volunteers without diabetes (Fig. 6A and B) and when cells from responders with DME were compared with those from nonresponders with DME (Fig. 6C and D). In particular, iPSC-ECs from nonresponders had significantly higher cell impedance (represented as a cell index) shortly after VEGF was added (Fig. 6C and D). At 30 and even 90 min after exposure to VEGF, iPSC-ECs derived from the groups with DME showed decreased cell impedance (i.e., increased permeability), and none showed a recovery pattern like that observed in healthy volunteers (Fig. 6A and B). iPSC-ECs from nonresponders showed significantly lower impedance than was seen in responders (Fig. 6C and D). We validated these data by performing further permeability experiments using three more iPSC-ECs lines (a total of six for each group), which confirmed increased FITC-dextran leakage in nonresponders (Supplementary Fig. 4). The relative percentage increase in FITC-dextran diffusion in nonresponders validated the cell impedance data mentioned above. Importantly, we compared the barrier integrity of our iPSC-ECs with that of primary human retinal microvascular ECs (HRMECs) using the xCELLigence system. The results showed that the iPSC-ECs were able to reach a level of impedance (cell index; proportional to barrier integrity) that was comparable to that of the HRMECs (data not shown). In addition, the obtained cell index values were comparable to or higher than those obtained by other groups working with human brain microvascular ECs (21). We can therefore confirm that our cells’ barrier integrity matches that of central nervous system ECs.

Figure 5

iPSC-ECs from donors without diabetes, patients with diabetes with mild DR, and patients with DME (both responders and nonresponders) have similar functional capacities. A and D: Representative images of an in vitro wound healing process at various time points for patients with diabetes and donors without diabetes (A) and for patients with DME (responders and nonresponders) (D). B and E: The migration capacity of iPSC-ECs is not significantly different between donors without diabetes and patients with diabetes (B) and among patients with DME (responders and nonresponders) (E). This capacity is measured as a chance in area from baseline according to a wound healing assay or scratch assay. Data are means ± SD (n = 3). C and F: iPSC-ECs from donors without diabetes and patients with diabetes (C) and among patients with DME (responders and nonresponders) (F) show no significant difference in proliferation capacity. Data are means ± SD (n = 3).

Figure 5

iPSC-ECs from donors without diabetes, patients with diabetes with mild DR, and patients with DME (both responders and nonresponders) have similar functional capacities. A and D: Representative images of an in vitro wound healing process at various time points for patients with diabetes and donors without diabetes (A) and for patients with DME (responders and nonresponders) (D). B and E: The migration capacity of iPSC-ECs is not significantly different between donors without diabetes and patients with diabetes (B) and among patients with DME (responders and nonresponders) (E). This capacity is measured as a chance in area from baseline according to a wound healing assay or scratch assay. Data are means ± SD (n = 3). C and F: iPSC-ECs from donors without diabetes and patients with diabetes (C) and among patients with DME (responders and nonresponders) (F) show no significant difference in proliferation capacity. Data are means ± SD (n = 3).

Close modal
Figure 6

Permeability response to VEGF stimulation was significantly different in iPSC-ECs from donors without diabetes and patients with diabetes (A and B), and in those from patients with DME (responders and nonresponders) (C and D). iPSC-ECs from nonresponders with DME had a significantly higher cell impedance shortly after the addition of VEGF compared to those of responders but a significantly lower impedance than in full responders after 90 minutes of VEGF treatment. Data are means ± SD (n = 3). *P<0.05, ***P < 0.001, ****P < 0.0001.

Figure 6

Permeability response to VEGF stimulation was significantly different in iPSC-ECs from donors without diabetes and patients with diabetes (A and B), and in those from patients with DME (responders and nonresponders) (C and D). iPSC-ECs from nonresponders with DME had a significantly higher cell impedance shortly after the addition of VEGF compared to those of responders but a significantly lower impedance than in full responders after 90 minutes of VEGF treatment. Data are means ± SD (n = 3). *P<0.05, ***P < 0.001, ****P < 0.0001.

Close modal

Phosphorylation Cascade Upon VEGF Stimulation Reveals Differences Between Responders and Nonresponders With DME

Given that significant differences were identified in permeability responses of iPSC-ECs from responders and nonresponders with DME, we aimed to shed light on the mechanism by which this might happen. VEGF induction of EC permeability starts with the binding of VEGF to VEGFR2, which then is phosphorylated, triggering a complex signaling cascade that culminates in the opening of adherens and tight junctions between ECs. VEGFR2 phosphorylation was assessed by stimulating iPSC-ECs from responders and nonresponders with VEGF for 5 and 20 min. Untreated iPSC-ECs from both groups were used as controls (0 min of VEGF stimulation). Interestingly, Western blotting has shown phosphorylation of VEGFR2 that was higher at the 5-min time point in samples from nonresponders than in responders (Fig. 7A and B). As a readout for downstream VEGFR2 signaling, phosphorylation of endothelial nitric oxide synthase (eNOS), which has a role in the regulation of endothelial permeability, was evaluated; no significant difference was found between groups (Fig. 7A and B). These results indicate that ECs generated from nonresponders have an increased phosphorylation pattern from VEGFR2.

Figure 7

Phosphorylation of proteins in response to VEGF stimulation in iPSC-ECs. A: Western blotting of phosphorylated VEGFR2 (P VEGFR2) in iPSC-ECs from responders and nonresponders with DME at various time points after VEGF treatment. β-actin was used as the loading control. P eNOS, phosphorylated eNOS. B: Quantification of the Western blotting. The phosphorylated VEGFR2 (Phospho-VEGFR2) signal at 5 min in nonresponders is double that in responders, whereas phosphorylated eNOS (Phospho-eNOS) shows no significant difference between the two groups. Data are means ± SD (n = 3). *P < 0.05.

Figure 7

Phosphorylation of proteins in response to VEGF stimulation in iPSC-ECs. A: Western blotting of phosphorylated VEGFR2 (P VEGFR2) in iPSC-ECs from responders and nonresponders with DME at various time points after VEGF treatment. β-actin was used as the loading control. P eNOS, phosphorylated eNOS. B: Quantification of the Western blotting. The phosphorylated VEGFR2 (Phospho-VEGFR2) signal at 5 min in nonresponders is double that in responders, whereas phosphorylated eNOS (Phospho-eNOS) shows no significant difference between the two groups. Data are means ± SD (n = 3). *P < 0.05.

Close modal

Differential mRNA Expression Between Responders and Nonresponders With DME and the Role of Neuronal Pentraxin 2

In order to obtain a more thorough analysis of the intrinsic differences between iPSC-ECs from responders and nonresponders, RNA sequencing was performed. This sequencing disclosed 13 DEGs (P <0.05) between responders and nonresponders (Fig. 8A and B). Further analysis revealed that these genes were involved in processes such as cell regeneration, repair, and survival (those showing higher expression in responders); and inflammation or susceptibility to diabetes (those showing higher expression in nonresponders). These 13 DEGs, of which 6 were upregulated in iPSC-ECs from responders (Fig. 8C) and 7 were upregulated in iPSC-ECs from nonresponders (Fig. 8D), could cluster biological replicates of similar samples together. Among these DEGs, neuronal pentraxin 2 (NPTX2) was particularly notable because it has been previously identified as a biomarker of edema in gliomas (22) and is also expressed in the retina (23). Strikingly, NPTX2 is also related to C-reactive protein, one of the inflammatory biomarkers involved in the pathogenesis of DR (24), which could mean that it contributes to inflammation in the microvasculature in nonresponders. The upregulation of NPTX2 was further assessed in iPSC-ECs from the different groups/cohorts by using quantitative RT-PCR (Fig. 8E). iPSC-ECs from nonresponders showed higher expression of NPTX2 than did those from responders. Because phosphorylated VEGFR2 was also higher in nonresponders after 5 min of exposure to VEGF (Fig. 7A), we hypothesized that a direct correlation may exist with NPTX2. As such, knockdown of NPTX2 for 72 h by lentiviral gene transfer in nonreponder iPSC-ECs showed significantly less phosphorylation of VEGFR2 at the 5-min time point than did the corresponding shNT control (Fig. 8F–H). In addition, given that NPTX2 is higher in nonresponders, VEGF levels might directly or indirectly regulate the level of NPTX2, as indicated in Fig. 8F. Further NPTX2 silencing experiments based on three additional patient-specific iPSC-EC lines from nonresponders showed a pattern of relative percentage decrease in diffusion of FITC-dextran in shNPTX2 (Supplementary Fig. 5). This could suggest that NPTX2 silencing might serve as a target for improving leakage in nonresponders.

Figure 8

Differences in RNA expression between responders and nonresponders with DME and the role of NPTX2. A: Volcano plot showing DEGs between responders and nonresponders. The x-axis represents the log2 fold change (Log2 FC), while the y-axis corresponds to −log10 adjusted P value (P & Log2FC); the significance cutoff of 1.3 is represented by the horizontal dotted line (equal to an adjusted P value of 0.05). Every red point in the plot corresponds to a significant DEG with a fold change greater than +1 for upregulated genes (right vertical dotted line) and less than −1 for downregulated genes (left vertical dotted line) for responders vs. nonresponders. NPTX2 is identified within the plot. B: A heat map cluster of the samples based on the 13 significant DEGs between responders and nonresponders. C: Relative expression of DEGs between responders and nonresponders. D and E: NPTX2 is upregulated in nonresponders. F: NPTX2 knockdown 72 h after lentiviral gene transfer in nonresponders delays phosphorylation of VEGFR2 at the 5-min time point after VEGF exposure. P VEGFR2, phosphorylated VEGFR2. G and H: Densitometry shows quantification of NPTX2 and phosphorylated VEGFR2 (P-VEGFR2). Data are means ± SD (n = 3). *P < 0.05, **P < 0.01.

Figure 8

Differences in RNA expression between responders and nonresponders with DME and the role of NPTX2. A: Volcano plot showing DEGs between responders and nonresponders. The x-axis represents the log2 fold change (Log2 FC), while the y-axis corresponds to −log10 adjusted P value (P & Log2FC); the significance cutoff of 1.3 is represented by the horizontal dotted line (equal to an adjusted P value of 0.05). Every red point in the plot corresponds to a significant DEG with a fold change greater than +1 for upregulated genes (right vertical dotted line) and less than −1 for downregulated genes (left vertical dotted line) for responders vs. nonresponders. NPTX2 is identified within the plot. B: A heat map cluster of the samples based on the 13 significant DEGs between responders and nonresponders. C: Relative expression of DEGs between responders and nonresponders. D and E: NPTX2 is upregulated in nonresponders. F: NPTX2 knockdown 72 h after lentiviral gene transfer in nonresponders delays phosphorylation of VEGFR2 at the 5-min time point after VEGF exposure. P VEGFR2, phosphorylated VEGFR2. G and H: Densitometry shows quantification of NPTX2 and phosphorylated VEGFR2 (P-VEGFR2). Data are means ± SD (n = 3). *P < 0.05, **P < 0.01.

Close modal

This study shows that only a proportion of patients with DME achieve full resolution of their macular fluid during the 1st year of treatment with anti-VEGF therapy. After first characterizing the phenotype of patients with DME and their responsiveness to anti-VEGF therapy, we demonstrated functional differences in their iPSC-ECs in vitro, which is to our knowledge a novel discovery. Importantly, the ability to separate patients with DME into categories of “full responders” and “nonresponders” to anti-VEGFs in a laboratory setting creates a unique platform for deeper patient stratification and for the understanding of pathogenic mechanisms of disease and response to anti-VEGF treatment.

Previous studies used CRT as seen on SD-OCT scans to determine the response to anti-VEGF therapy. No consensus exists, however, on definitions of “responder” and “nonresponder.” Thus, Lee et al. (13) considered a response to have occurred when a CRT was reduced by >50 µm after three consecutive anti-VEGF injections, whereas Dabir et al. (14) determined a response when CRT was reduced by >10% after two consecutive injections. Pieramici et al. (11), using data from RISE and RIDE (A Study of Ranibizumab Injection in Subjects With Clinically Significant Macular Edema [ME] With Center Involvement Secondary to Diabetes Mellitus), proposed the definition of “immediate” and “delayed” responders based on whether a reduction in CRT of >10% or ≤10%, respectively, had occurred following three loading doses of anti-VEGF. Based on data from the DRCR.net protocol I, Dugel et al. (12) used the terms “strong early” and “limited early” responders for a reduction of ≥20% or <20%, respectively, of CRT on SD-OCT after the same loading doses. CRT as measured on SD-OCT scans, though helpful, may not be the best way to determine the response to anti-VEGF therapy given the great range of normal values among individuals, variability due to the instruments used in order to obtain CRT values, and the fact that DME may still be present in the context of a “normal” CRT value in patients in whom neurodegeneration may have occurred. We considered “full responders” to be those individuals who achieved a fully dry macula at any time during the 1st year of treatment. In our cohort, the macula of full responders dried after a median of 7 months (range, 1–12 months). Waiting longer than a year to determine a response to treatment and to consider switching to an alternative therapy may compromise functional recovery. In only a small percentage of patients in our study did the macula dry fully after a year of adequate treatment with an anti-VEGFs.

Although cost-effectiveness has been demonstrated for anti-VEGF therapies licensed for the treatment of DME and currently in use in clinical practice—ranibizumab [25] and aflibercept [26]—the data used to evaluate their clinical effectiveness and cost-effectiveness was obtained from RCTs. It is now acknowledged, however, that RCT results often are not reproducible in clinical practice (27). Thus, whether the cost-effectiveness of anti-VEGF therapies remains above appropriate thresholds in real-life clinical settings remains to be elucidated.

In this study we used iPSC technology to make an in vitro model of the retinal microvasculature of patients with DME. In most previous diabetes-related studies, the main source for iPSC generation was dermal fibroblasts (28). These cells are easily maintained and reprogrammed but must be obtained through a skin biopsy (29), a procedure that carries ethical challenges, especially for patients with diabetes, in whom healing can be delayed (30). In contrast, we demonstrated that iPSCs can be obtained successfully from blood MNCs by using nonintegrating episomal-based transfection and by avoiding the long-term incorporation of reprogramming factors into chromosomal DNA.

Patient-specific iPSCs were differentiated into vascular ECs, as they are critical components of the retinal neurovascular unit (NVU) and their dysfunction is intimately linked to a loss of integrity of the inner BRB. Primary ECs have been widely used to study endothelial permeability and molecular pathways in vitro (31,32), although they are often obtained from animals or from human donors after death, and the human disease–related information they can provide is limited. In contrast, iPSC-ECs generated in this study provided a human disease model that, importantly, is specifically linked to individual patients who have been clinically phenotyped. In addition, because iPSCs can retain information from the donor cells in terms of epigenetic memory (33), the subsequent differentiated cells may be predisposed to exhibit a donor-related epigenetic signature. One study recently reported that vascular progenitors generated from tankyrase inhibitor–regulated naive diabetic human iPSCs revascularized the ischemic retina, suggesting erasure of dysfunctional epigenetic donor-cell memory (34). This phenomenon also has been described in other cells such as cardiomyocytes, wherein treatment with large amounts of glucose induces “metabolic memory” (35).

iPSC-ECs, as generated herein, are not retina-specific. However, one of the key characteristics of retinal microvascular ECs is their low permeability, which is due to their tight barrier properties. Even if the iPSC-ECs used in this study are not retina-specific, their barrier properties, checked as cell impedance, are equivalent to those of HRMECs and human brain microvascular ECs (21). Furthermore, although ECs are key to the pathogenesis of DME, they are not the only cells responsible for the maintenance of the inner BRB; integrity of the NVU is also influenced by pericytes, Müller glia, neurons, and immune cells. In contrast, the outer BRB is formed by the retinal pigment epithelium, which is also dysfunctional in diabetes (36). Considering that iPSCs can differentiate into any type of cell in the body, studies are currently underway in our laboratory to generate retinal-specific cells such as retinal pigment epithelial cells and Müller glia in order to evaluate their role in DME in a patient-specific manner (37). Cocultures of iPSC-ECs with other iPSC-derived cells that form the NVU (38) would also provide a model that more closely resembles the living retina (39).

In this study, iPSC-ECs from donors without diabetes, patients with diabetes and mild DR, and responders and nonresponders with DME displayed similar functional capacities. The migration capacity of iPSC-ECs was not significantly different between these groups. In a similar manner, no significant difference was found in their proliferation capacity. In particular, when iPSC-ECs from responders and nonresponders were compared, no significant differences were found in their morphology, expression of EC markers, and most functional capacities, according to the tests performed. This confirms that iPSCs from patients with diabetes can be efficiently differentiated into functional ECs.

Because excessive EC permeability exists in DME, we assessed the barrier function capacity of iPSC-ECs and their permeability response to VEGF in vitro. When analyzing iPSC-ECs from responders and nonresponders, we found that these cells did not recover their initial EC barrier function even 90 min after stimulation by VEGF. Interestingly, our results showed that iPSC-ECs from nonresponders had a higher permeability response to VEGF than did cells from responders, suggesting that ECs from nonresponders are more sensitive to the effects of VEGF. This suggests that ECs from nonresponders may be activated even with very low levels of intraocular VEGF, which may remain despite the blockade achieved by anti-VEGFs. Because the VEGF-mediated increase of EC permeability is driven by a complex phosphorylation cascade (40), we assessed the phosphorylation status of VEGFR2, eNOS, and 46 other kinases (Fig. 7 and Supplementary Fig. 3) after VEGF stimulation in iPSC-ECs from responders and nonresponders. We focused on several VEGF-related pathways. Results showed that VEGFR2 was differentially phosphorylated between these groups, with nonresponders demonstrating greater phosphorylation of this receptor than responders. These results are in agreement with those from the permeability studies, reinforcing the hypothesis that nonresponders may be highly sensitive to the effects induced by VEGF. If the nonresponder receptors were more sensitive to VEGF and therefore had a lower threshold for VEGFR2 activation, even the low levels of VEGF that may persist after anti-VEGF blockade in vivo would be able to maintain vascular leakage through VEGFR2 signaling, with DME persisting despite treatment.

In order to deepen our understanding of the differences between these two groups, RNA sequencing of their iPSC-ECs was performed. A differential gene expression profile was observed between responders and nonresponders. A total of 13 DEGs were identified as significantly expressed when the adjusted P value was <0.05 and the absolute log2 fold change was >1. One of the genes found to be upregulated in nonresponders was NPTX2, which has been identified as a potential biomarker of edema in gliomas (22) and seems to be related to C-reactive protein, one of the inflammatory biomarkers involved in the pathogenesis of DR (41). Thus higher expression of this protein in the endothelium of nonresponders could be driving edema through a VEGF parallel pathway, which could be an additional reason why these patients fail to respond to VEGF inhibition. Silencing NPTX2 in nonresponders diminished the effect of VEGF in the phosphorylation of VEGFR2, suggesting a possible effect in reducing inflammation-related events. Furthermore, NPTX2 silencing reduced the relative FITC-dextran leakage in nonresponders, showcasing that targeting this gene could play a role in correcting barrier integrity. Studies have shown that abnormal upregulation of NPTX2 expression was linked to the proliferation and metastasis of cancers such as clear-cell renal cell carcinoma through the activation of the Wnt/β-catenin signaling pathway. NPTX2 was shown to interact with frizzled class receptor 6 to induce β-catenin nuclear translocation, with an ensuing decrease in the expression of E-cadherin (42) and an increase in the expression of MYC, snail, N-cadherin, and cyclin D1, which induces VEGF production (43).

The strengths of this study include the identification of consecutive patients through an electronic database; the masking of those who assessed outcomes (i.e., the researcher who graded the OCT scans and, very importantly, the scientists undertaking the iPSC-EC experiments and analyzing their results, who were masked to the treatment response observed in the clinics); the evaluation of all OCT scans obtained throughout the entire 1st year of treatment in order to determine the treatment response in all patients included in the cohort; and the robust techniques used in order to obtain patient-specific iPSC and iPSC-ECs. One of the limitations is the small sample size (n = 6) used for the in vitro studies. However, because these patient-specific iPSC-ECs were generated from a very well-characterized clinical cohort, they revealed robust differences and allowed conclusions to be made. This study sets a foundation for identifying key pathophysiological differences between full responders and nonresponders to anti-VEGF treatment among patients with DME, which may have potential clinical relevance, and provides a scope for new drug development.

Conclusions

By generating iPSC-ECs from volunteers without diabetes, people with diabetes but no DME, and people with DME who were categorized as full responders or nonresponders to anti-VEGFs, we found increased cell permeability in those with diabetes after stimulation with VEGF in vitro—an observation that was exaggerated in nonresponders. Additionally, phosphorylation of key cellular proteins after exposure to VEGF, including VEGFR2, and gene expression profiles (e.g., of NPTX2) differed between full responders and nonresponders, allowing for the possible identification in the future of patients who will respond to treatment.

This article contains supplementary material online at https://doi.org/10.2337/figshare.12732944.

Acknowledgments. The authors thank Eimear Byrne (Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, UK) for her expert advice on the FITC-dextran experiments.

Funding. This work was supported by grants from the Medical Research Council, the Biotechnology and Biological Sciences Research Council, and the British Heart Foundation. The authors also acknowledge the support of the Belfast Association for the Blind.

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

Author Contributions. M.V.G., M.E., and S.K. conceived and designed the study; collected, assembled, analyzed, and interpreted data; and wrote the manuscript. H.N.-M. analyzed and interpreted RNA sequencing data. S.F., S.S., and G.V. collected and assembled data. D.J.G. provided study material and approved the final manuscript. A.W.S., N.L., and A.M. conceived and designed the study; collected, assembled, analyzed, and interpreted data; wrote the manuscript; obtained financial support; and approved the final manuscript. N.L. and A.M. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Lee
R
,
Wong
TY
,
Sabanayagam
C
.
Epidemiology of diabetic retinopathy, diabetic macular edema and related vision loss
.
Eye Vis (Lond)
2015
;
2
:
17
2.
Cunha-Vaz
J
,
Faria de Abreu
JR
,
Campos
AJ
.
Early breakdown of the blood-retinal barrier in diabetes
.
Br J Ophthalmol
1975
;
59
:
649
656
3.
Stitt
AW
,
Curtis
TM
,
Chen
M
, et al
.
The progress in understanding and treatment of diabetic retinopathy
.
Prog Retin Eye Res
2016
;
51
:
156
186
4.
Funatsu
H
,
Yamashita
H
,
Nakamura
S
, et al
.
Vitreous levels of pigment epithelium-derived factor and vascular endothelial growth factor are related to diabetic macular edema
.
Ophthalmology
2006
;
113
:
294
301
5.
Díaz-Coránguez
M
,
Ramos
C
,
Antonetti
DA
.
The inner blood-retinal barrier: cellular basis and development
.
Vision Res
2017
;
139
:
123
137
6.
Elman
MJ
,
Aiello
LP
,
Beck
RW
,et al.;
Diabetic Retinopathy Clinical Research Network
.
Randomized trial evaluating ranibizumab plus prompt or deferred laser or triamcinolone plus prompt laser for diabetic macular edema
.
Ophthalmology
2010
;
117
:
1064
1077.e35
7.
Nguyen
QD
,
Brown
DM
,
Marcus
DM
, et al.;
RISE and RIDE Research Group
.
Ranibizumab for diabetic macular edema: results from 2 phase III randomized trials: RISE and RIDE
.
Ophthalmology
2012
;
119
:
789
801
8.
Mitchell
P
,
Bandello
F
,
Schmidt-Erfurth
U
, et al.;
RESTORE study group
.
The RESTORE study: ranibizumab monotherapy or combined with laser versus laser monotherapy for diabetic macular edema
.
Ophthalmology
2011
;
118
:
615
625
9.
Korobelnik
JF
,
Do
DV
,
Schmidt-Erfurth
U
, et al
.
Intravitreal aflibercept for diabetic macular edema
.
Ophthalmology
2014
;
121
:
2247
2254
10.
Wells
JA
,
Glassman
AR
,
Ayala
AR
, et al.;
Diabetic Retinopathy Clinical Research Network
.
Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema
.
N Engl J Med
2015
;
372
:
1193
1203
11.
Pieramici
DJ
,
Wang
PW
,
Ding
B
,
Gune
S
.
Visual and anatomic outcomes in patients with diabetic macular edema with limited initial anatomic response to ranibizumab in RIDE and RISE
.
Ophthalmology
2016
;
123
:
1345
1350
12.
Dugel
PU
,
Campbell
JH
,
Kiss
S
, et al
.
Association between early anatomic response to anti-vascular endothelial growth factor therapy and long-term outcome in diabetic macular edema: an independent analysis of protocol i study data
.
Retina
2019
;
39
:
88
97
13.
Lee
J
,
Moon
BG
,
Cho
AR
,
Yoon
YH
.
Optical coherence tomography angiography of DME and its association with anti-VEGF treatment response
.
Ophthalmology
2016
;
123
:
2368
2375
14.
Dabir
SS
,
Das
D
,
Nallathambi
J
,
Mangalesh
S
,
Yadav
NK
,
Schouten
JS
.
Differential systemic gene expression profile in patients with diabetic macular edema: responders versus nonresponders to standard treatment
.
Indian J Ophthalmol
2014
;
62
:
66
73
15.
Elman
MJ
,
Ayala
A
,
Bressler
NM
, et al.;
Diabetic Retinopathy Clinical Research Network
.
Intravitreal Ranibizumab for diabetic macular edema with prompt versus deferred laser treatment: 5-year randomized trial results
.
Ophthalmology
2015
;
122
:
375
381
16.
Sachdeva
MM
,
Moshiri
A
,
Leder
HA
,
Scott
AW
.
Endophthalmitis following intravitreal injection of anti-VEGF agents: long-term outcomes and the identification of unusual micro-organisms
.
J Ophthalmic Inflamm Infect
2016
;
6
:
2
17.
Vilà-González
M
,
Kelaini
S
,
Magee
C
, et al
.
Enhanced function of induced pluripotent stem cell-derived endothelial cells through ESM1 signaling
.
Stem Cells
2019
;
37
:
226
239
18.
Margariti
A
,
Zampetaki
A
,
Xiao
Q
, et al
.
Histone deacetylase 7 controls endothelial cell growth through modulation of beta-catenin
.
Circ Res
2010
;
106
:
1202
1211
19.
Margariti
A
,
Winkler
B
,
Karamariti
E
, et al
.
Direct reprogramming of fibroblasts into endothelial cells capable of angiogenesis and reendothelialization in tissue-engineered vessels
.
Proc Natl Acad Sci U S A
2012
;
109
:
13793
13798
20.
Cochrane
A
,
Kelaini
S
,
Tsifaki
M
, et al
.
Quaking is a key regulator of endothelial cell differentiation, neovascularization, and angiogenesis
.
Stem Cells
2017
;
35
:
952
966
21.
Wiltshire
R
,
Nelson
V
,
Kho
DT
,
Angel
CE
,
O’Carroll
SJ
,
Graham
ES
.
Regulation of human cerebro-microvascular endothelial baso-lateral adhesion and barrier function by S1P through dual involvement of S1P1 and S1P2 receptors
.
Sci Rep
2016
;
6
:
19814
22.
Carlson
MR
,
Pope
WB
,
Horvath
S
, et al
.
Relationship between survival and edema in malignant gliomas: role of vascular endothelial growth factor and neuronal pentraxin 2
.
Clin Cancer Res
2007
;
13
:
2592
2598
23.
Bjartmar
L
,
Huberman
AD
,
Ullian
EM
, et al
.
Neuronal pentraxins mediate synaptic refinement in the developing visual system
.
J Neurosci
2006
;
26
:
6269
6281
24.
Nowak
M
,
Wielkoszyński
T
,
Marek
B
, et al
.
Antioxidant potential, paraoxonase 1, ceruloplasmin activity and C-reactive protein concentration in diabetic retinopathy
.
Clin Exp Med
2010
;
10
:
185
192
25.
National Institute for Health and Care Excellence
.
Ranibizumab for treating diabetic macular oedema. Technology Appraisal Guidance, 2013
.
Accessed 29 April 2020. Available from https://www.nice.org.uk/guidance/ta274
26.
National Institute for Health and Care Excellence
.
Aflibercept for treating diabetic macular oedema. Technology Appraisal Guidance, 2015
.
Accessed 29 April 2020. Available from https://www.nice.org.uk/guidance/ta346
27.
Denniston
AK
,
Lee
AY
,
Lee
CS
, et al.;
UK DR EMR Users Group
.
United Kingdom Diabetic Retinopathy Electronic Medical Record (UK DR EMR) Users Group: report 4, real-world data on the impact of deprivation on the presentation of diabetic eye disease at hospital services
.
Br J Ophthalmol
2019
;
103
:
837
843
28.
Kondo
Y
,
Toyoda
T
,
Inagaki
N
,
Osafune
K
.
iPSC technology-based regenerative therapy for diabetes
.
J Diabetes Investig
2018
;
9
:
234
243
29.
Vangipuram
M
,
Ting
D
,
Kim
S
,
Diaz
R
,
Schüle
B
.
Skin punch biopsy explant culture for derivation of primary human fibroblasts
.
J Vis Exp
2013
;(
77
):
e3779
30.
Dangwal
S
,
Stratmann
B
,
Bang
C
, et al
.
Impairment of wound healing in patients with type 2 diabetes mellitus influences circulating microRNA patterns via inflammatory cytokines
.
Arterioscler Thromb Vasc Biol
2015
;
35
:
1480
1488
31.
van Dijk
IA
,
Ferrando
ML
,
van der Wijk
AE
, et al
.
Human salivary peptide histatin-1 stimulates epithelial and endothelial cell adhesion and barrier function
.
FASEB J
2017
;
31
:
3922
3933
32.
Imai
A
,
Toriyama
Y
,
Iesato
Y
, et al
.
Adrenomedullin suppresses vascular endothelial growth factor-induced vascular hyperpermeability and inflammation in retinopathy
.
Am J Pathol
2017
;
187
:
999
1015
33.
Kim
K
,
Doi
A
,
Wen
B
, et al
.
Epigenetic memory in induced pluripotent stem cells
.
Nature
2010
;
467
:
285
290
34.
Park
TS
,
Zimmerlin
L
,
Evans-Moses
R
, et al
.
Vascular progenitors generated from tankyrase inhibitor-regulated naïve diabetic human iPSC potentiate efficient revascularization of ischemic retina
.
Nat Commun
2020
;
11
:
1195
35.
Yu
XY
,
Geng
YJ
,
Liang
JL
, et al
.
High levels of glucose induce “metabolic memory” in cardiomyocyte via epigenetic histone H3 lysine 9 methylation
.
Mol Biol Rep
2012
;
39
:
8891
8898
36.
Duh
EJ
,
Sun
JK
,
Stitt
AW
.
Diabetic retinopathy: current understanding, mechanisms, and treatment strategies
.
JCI Insight
2017
;
2
:
e93751
37.
Yang
J
,
Cai
B
,
Glencer
P
,
Li
Z
,
Zhang
X
,
Li
X
.
Induced pluripotent stem cells and outer retinal disease
.
Stem Cells Int
2016
;
2016
:
2850873
38.
Orlova
VV
,
Drabsch
Y
,
Freund
C
, et al
.
Functionality of endothelial cells and pericytes from human pluripotent stem cells demonstrated in cultured vascular plexus and zebrafish xenografts
.
Arterioscler Thromb Vasc Biol
2014
;
34
:
177
186
39.
Hollmann
EK
,
Bailey
AK
,
Potharazu
AV
,
Neely
MD
,
Bowman
AB
,
Lippmann
ES
.
Accelerated differentiation of human induced pluripotent stem cells to blood-brain barrier endothelial cells
.
Fluids Barriers CNS
2017
;
14
:
9
40.
Weis
SM
,
Cheresh
DA
.
Pathophysiological consequences of VEGF-induced vascular permeability
.
Nature
2005
;
437
:
497
504
41.
Lim
LS
,
Tai
ES
,
Mitchell
P
, et al
.
C-reactive protein, body mass index, and diabetic retinopathy
.
Invest Ophthalmol Vis Sci
2010
;
51
:
4458
4463
42.
Xu
C
,
Tian
G
,
Jiang
C
, et al
.
NPTX2 promotes colorectal cancer growth and liver metastasis by the activation of the canonical Wnt/β-catenin pathway via FZD6
.
Cell Death Dis
2019
;
10
:
217
43.
Tsai
HL
,
Yeh
YS
,
Chang
YT
, et al
.
Co-existence of cyclin D1 and vascular endothelial growth factor protein expression is a poor prognostic factor for UICC stage I-III colorectal cancer patients after curative resection
.
J Surg Oncol
2013
;
107
:
148
154
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/content/license.