Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 3.
Published in final edited form as: Methods Mol Biol. 2022;2416:133–156. doi: 10.1007/978-1-0716-1908-7_10

Generation of Pericytic-Vascular Progenitors from Tankyrase/PARP-Inhibitor-Regulated Naïve (TIRN) Human Pluripotent Stem Cells

Ludovic Zimmerlin 1, Tea Soon Park 1, Imran Bhutto 1, Gerard Lutty 1, Elias T Zambidis 1
PMCID: PMC9529319  NIHMSID: NIHMS1837983  PMID: 34870835

Abstract

Tankyrase/PARP inhibitor-regulated naïve human pluripotent stem cells (TIRN-hPSC) represent a new class of human stem cells for regenerative medicine that can differentiate into multi-lineage progenitors with improved in vivo functionality. Chemical reversion of conventional, primed hPSC to a TIRN-hPSC state alleviates dysfunctional epigenetic donor cell memory, lineage-primed gene expression, and potentially disease-associated aberrations in their differentiated progeny. Here, we provide methods for the reversion of normal or diseased patient-specific primed hPSC to TIRN-hPSC and describe their subsequent differentiation into embryonic-like pericytic-endothelial “naïve” vascular progenitors (N-VP). N-VP possess improved vascular functionality, high epigenetic plasticity, maintain greater genomic stability, and are more efficient in migrating to and re-vascularizing ischemic tissues than those generated from primed isogenic hPSC. We also describe detailed methods for the ocular transplantation and quantitation of vascular engraftment of N-VP into the ischemia-damaged neural retina of a humanized mouse model of ischemic retinopathy. The application of TIRN-hPSC-derived N-VP will advance vascular cell therapies of ischemic retinopathy, myocardial infarction, and cerebral vascular stroke.

Keywords: Naïve pluripotency, Vascular regeneration, Vascular progenitors, Pericyte, Human pluripotent stem cell, Differentiation, Tankyrase inhibition, PARP, Ischemic retinopathy

1. Introduction

Human vasculature arises during embryonic development at extraembryonic and embryonic locations from highly proliferative mesoderm-derived vascular progenitors (VP). VP generate pericytes and endothelial progenitor cells and form interconnected primary vitelline, embryonic, and placental circulations [1]. Embryonic-like VP, defined by the cell-surface markers CD31+ CD146+ CXCR4+ can similarly be generated from human embryonic stem cell (hESC) and human induced pluripotent stem cell (hiPSC), and appear to functionally phenocopy the mesodermal lineage that gives rise to embryonic human vasculature [27]. Such human pluripotent stem cell (hPSC)-derived VP generated endothelial (CD31+ CD34+ CD144+ CD105+) and pericytic (CD140b+ CD105lo) populations (Fig. 1a, b) with highly efficient blood vessel-forming abilities (Fig. 1cf) that regenerated ischemia-damaged adult retinal blood vessels in a humanized murine ischemia/reperfusion (I/R) model [2, 3].

Fig. 1.

Fig. 1

Generation of embryonic pericytic-endothelial VP from primed hPSC. (a) Primed H9 hESC were differentiated to vascular mesoderm. CD31+ CD146+ CXCR4+ VP were further FACS-purified (red gate), and culture-expanded in EGM-2 medium into populations that contained distinct (b) endothelial CD144+ CD105+ CD34+ (purple-gated) and pericytic CD140b+ CD34 CD105lo (green-gated) populations. Expanded VP were assayed for endothelial function with (c) Ac-Dil-LDL uptake assays, and (d) In vivo Matrigel plug chimeric vascular network formation in NOD/SCID mice. Sections were stained with anti-human CD31 antibodies (brown). (e) Primed cord blood-hiPSC-derived CD31+ CD146+ VP formed assembled vascular tubes in collagen gel. (f) These tubes were imaged with transmission electron microscopy, which revealed cooperating endothelial-pericytic-like cells with bifurcations. L: lumen, n: nuclei

Although multipotent embryonic-like VP may offer patient-specific vascular therapies [811], such VP with regenerative capacities are rare and limited in proliferative expansion in the adult, especially in diabetics [1217]. Furthermore, primed hPSC lines remain restricted in their ultimate utility for cell therapies by highly variable vascular differentiation efficiencies [3, 6, 18, 19] and poor in vivo functionality of the VP derived from them [2, 3]. This limitation is ultimately due to the developmental similarities of hPSC with primed murine post-implantation epiblast stem cells (mEpiSC) which adopt a spectrum of pluripotent states with highly variable lineage-primed transcriptomes and post-implantation epiblast epigenetic marks [6, 7, 20]. As a result, primed hPSC possess a restricted multi-lineage differentiation potential compared to naïve epiblast-like ESC [20]. Moreover, epigenetic aberrations in diseased states, such as diabetes, inhibit the efficient somatic donor cell reprogramming to a functional primed pluripotent state [1214, 21, 22].

Naïve-like hPSC with pre-implantation epiblast phenotypes, decreased lineage priming, improved epigenomic stability, and higher functionality of differentiated progenitors may solve these obstacles. Several groups have reported various small molecule approaches that putatively captured human naïve epiblast-like pluripotent states that were more primitive than those exhibited by primed hPSC (reviewed in [20]). However, many of these human naïve states exhibited karyotypic instability, global loss of parental genomic imprinting, and impaired multi-lineage differentiation performance that was attributed to the small molecule cocktails that were employed. In contrast, Zimmerlin and colleagues demonstrated that these caveats could be avoided by culturing primed hESC and hiPSC in a cocktail termed “LIF-3i” comprising of the small molecule tankyrase/PARP (poly ADP ribose polymerase) inhibitor XAV939, the GSK3β inhibitor CHIR99021, the MEK inhibitor PD0325901, and LIF [6, 7].

The LIF-3i method rapidly reverted primed, conventional hiPSC and hESC to a stable Tankyrase/PARP inhibitor-regulated naïve hPSC (TIRN-hPSC) state that adopted biochemical, transcriptional, and epigenetic features of the human pre-implantation naïve epiblast. TIRN reversion conferred a broad repertoire of normal and diabetic conventional hiPSC with molecular characteristics unique to naïve pluripotency, including increased phosphorylated STAT3 signaling, decreased ERK phosphorylation, global 5-methylcytosine CpG hypomethylation, genome-wide CpG demethylation at ESC-specific gene promoters, and dominant distal POU5F1 (OCT4) enhancer usage. TIRN-hiPSC maintained normal karyotypes and were devoid of systematic loss of imprinted CpG patterns or irreversible demethylation defects reported in other naïve reversion systems that were attributed to prolonged culture with MEK inhibitors [6, 7].

More importantly, reversion of primed hPSC to TIRN-hPSC improved multi-lineage potency, eliminated interline variability of differentiation among primed hPSC, and, significantly, erased primed hPSC donor cell “epigenetic memory” (Fig. 2a) [2, 6, 7]. The improved functional pluripotency of TIRN-hPSC was directly potentiated by the inclusion of XAV939 to the classical 2i-based culture media. Interestingly, inclusion of XAV939 into other systems has now identified stem cells with expanded pluripotency capable of generating both embryonic and extra-embryonic lineages [23, 24]. Thus, TIRN-hPSC represent a new class of human stem cells with improved multi-lineage functionality and potentially high impact for regenerative medicine.

Fig. 2.

Fig. 2

(a) Waddington landscape model for TIRN reversion (red) of primed hiPSC (blue), adapted from [2]. Epigenetic obstacles to VP differentiation may be overcome with molecular reversion to a TIRN epiblast-like state (red) with a developmentally more primitive epigenetic configuration. (b) Schema of paradigm for generating cGMP-grade, HLA-defined TIRN-hiPSC (from reprogrammed CD34+ hematopoietic progenitors) with improved multi-lineage vascular, neural, and retinal pigmented epithelial (RPE) differentiation for comprehensive regeneration of ischemic neural retina

In a further advance of this human naïve stem cell system, Park and colleagues reported that the functionality of VP generated from both normal and diabetic primed hiPSC was significantly improved following reversion to a TIRN epiblast-like state [2]. Primed diabetic hiPSC were reprogrammed from type-1 diabetic donor fibroblasts and stably reverted to diabetic TIRN-hiPSC. Embryonic diabetic naïve VP (N-VP) differentiated from diabetic TIRN-hiPSC expanded more efficiently, possessed more stable genomic integrity, and displayed higher in vitro vascular functionality than VP generated from isogenic primed diabetic hiPSC. Moreover, human diabetic N-VP survived, migrated, and engrafted in vivo into the deep vasculature of the neural retinal layers with significantly higher efficiencies than isogenic primed diabetic VP in a humanized murine model of ischemic retinopathy.

Analyses of CpG DNA methylation and histone configurations at developmental promoters of diabetic TIRN-hPSC and diabetic N-VP revealed tight lineage-specific gene expression, and a de-repressed embryonic state with high epigenetic plasticity. TIRN-hPSC generated N-VP from a diseased state with improved epigenetic plasticity and superior vascular regenerative function [2, 6, 7]. Thus, reprogramming of patient donor cells to TIRN-hPSC may erase acquired epigenetic aberrations sustained from dysfunctional donor cell or disease-associated epigenetic memory (Fig. 2a) [2].

Here, we describe how to generate prolific human CD31+ CD146+ CXCR4+ N-VP from TIRN-hPSC, and detail the methods for transplanting these cells therapeutically in vivo into a humanized murine model of ischemic retinopathy. Endothelial-pericytic N-VP possess superior capacities for restoring blood flow to ischemia-damaged neural retina, relative to primed VP that are differentiated from conventional, primed hPSC. Clinical-grade, patient-specific or HLA-banked N-VP with improved functionalities could be used for regenerating, stabilizing, or even reversing end-stage blinding complications. Moreover, efficient differentiation of N-VP in parallel with neural retinal progenitors from the same TIRN-hiPSC line could comprehensively regenerate multiple cell types rendered defective in an adult diseased ocular environment [25, 26] (Fig. 2b). This N-VP system could be further optimized to establish a paradigm for future comprehensive multi-lineage vascular repair of other diseased ischemic tissues (e.g., cerebrovascular stroke and myocardial ischemia) using either patient-specific or banked HLA-defined TIRN-hiPSC [27, 28].

2. Materials

2.1. TIRN-hPSC Cell Culture and N-VP Differentiation

  1. Essential 8 (E8) medium (see Note 1): DMEM/F-12 with 2.5 mM L-Glutamine, 15 mM HEPES and 14 mM sodium bicarbonate, supplemented with 50–100 ng/mL recombinant human FGF2, 2 ng/mL recombinant human TGF-β1, 64 μg/mL L-ascorbic acid-2-phosphate magnesium, 14 ng/mL sodium selenite, 10.7 μg/mL recombinant human transferrin, and 20 μg/mL recombinant human insulin. Sterile filter. Store at 4 °C for up to 3 weeks. Prewarm before use.

  2. Vitronectin-XF solution (10 μg/mL).

  3. Versene or phosphate-buffered saline (PBS)-based enzyme-free cell dissociation buffer.

  4. LIF-3i TIRN medium (see Note 2): DMEM/F-12 with GlutaMAX, supplemented with 20% KnockOut Serum Replacement (KOSR), 0.1 mM MEM non-essential amino acids, 1 mM L-Glutamine, 0.1 mM β-mercaptoethanol, 20 ng/mL recombinant human LIF, 3 μM CHIR99021, 1 μM PD0325901, and 4 μM XAV939. Sterile filter the complete medium. Store at 4 °C for up to 3 weeks. Prewarm before use.

  5. LIF-5i TIRN adaptation medium: supplement LIF-3i TIRN medium with 10 μM Forskolin, 2 μM purmorphamine, and 10 ng/mL recombinant human FGF2. Sterile filter. Store at 4 °C for up to 3 weeks. Prewarm before use.

  6. Murine embryonic fibroblast (MEF) medium: DMEM supplemented with 10% heat-inactivated fetal bovine serum (FBS), 1× MEM non-essential amino acids, 0.1 mM β-mercaptoethanol, 1 mM L-glutamine and 0.5% penicillin/streptomycin. Sterile filter the complete medium.

  7. LIF-3i/5i TIRN hESC and hiPSC plate preparation (see Note 3): 6-well culture-treated plates pre-coated overnight with sterile 0.1% gelatin solution, and pre-seeded with mitotically inactive MEF. Rinse wells twice with PBS just before use.

  8. Accutase.

  9. TIRN cryopreservation medium (10% DMSO, 40% KOSR, 50% TIRN medium, and 5 μM Y-27632).

  10. Vascular mesoderm specification medium (see Note 4): Sterile-filter Albumin Polyvinyl-alcohol Essential Lipids (APEL) low insulin medium [29] and supplement with 25 ng/mL human Activin A, 50 ng/mL human VEGF165, 30 ng/mL human BMP4, and 1.5 μM CHIR99021. Store at 4 °C for up to 3 weeks. Prewarm before use.

  11. N-VP differentiation medium (see Note 4): supplement sterile-filtered APEL low insulin medium with 50 ng/mL human VEGF and 10 μM SB431542. Store at 4 °C for up to 3 weeks. Prewarm before use.

  12. Magnetic activated cell sorting (MACS) system for N-VP isolation (see Note 5): human CD31 microbead kit, LS columns, and MidiMACS separator. MACS wash buffer consists of PBS supplemented with 0.04% Bovine Serum Albumin (BSA).

  13. N-VP expansion medium: supplement endothelial growth medium-2 (EGM-2; commercially available from Lonza) with an additional 25 ng/mL of human VEGF165. Store at 4 °C for up to 3 weeks. Prewarm before use.

  14. N-VP expansion plate preparation: 6-well culture-treated plates coated overnight with 10 μg/mL fibronectin solution.

2.2. Ocular Ischemic Reperfusion (I/R) Injury Mouse Model Reagents

  1. 6–8-week-old male NOD/Shi-scid/IL-2Rγnull (NOG) mice (see Note 6).

  2. Deep anesthesia injectable solution: 0.9% sodium chloride solution, 50 mg/kg ketamine, 10 mg/kg xylazine. Anesthesia is maintained by two additional 50 μL intramuscular injections of 20 mg/mL ketamine solution.

  3. Animal pupil dilation solution: 2.5% phenylephrine hydrochloride ophthalmic solution.

  4. Ophthalmic topical anesthetic solution: 0.5% tetracaine hydrochloride.

  5. Anterior chamber cannulation (see Note 7): ophthalmic surgical microscope, 30-gauge needle, silicone infusion line, sterile irrigating balanced salt solution.

  6. Ophthalmic topical antibiotic ointment: Bacitracin zinc and Polymyxin B sulfate (AK-Poly-Bac).

  7. Digital pico-injector (PLI-100; purchase from Harvard Apparatus, Holliston, MA, USA).

2.3. Whole Mount Retinal Preparation and Immunostaining Reagents

  1. Tris-Buffered Saline (TBS), adjusted to pH 7.4 with HCl.

  2. Fixative solution (see Note 8): 2% paraformaldehyde in TBS.

  3. Permeabilization solution: 0.1% Triton X-100 in TBS.

  4. Blocking solution: TBS, 2% goat serum, 1% BSA.

  5. Antibodies (see Note 9): Cy3-conjugated anti-human nuclear antigen (HNA), rabbit anti-mouse Collagen IV, rat anti-mouse CD31, Cy3-conjugated goat anti-rabbit secondary antibody, and Alexafluor-647-conjugated goat anti-rat secondary antibody.

  6. Fluorescent nuclear dye (e.g., 4′,6-diamidino-2-phenylindole (DAPI) or Hoechst 33342).

  7. Glass slides and coverslips.

  8. Confocal microscope with 10× objective and tile-scan feature.

2.4. Cross-Section Retinal Preparation and Immunostaining Reagents

  1. Fixative solution: 0.1 M Sodium Phosphate, 2% paraformaldehyde, 5% sucrose, adjust to pH 7.2 with HCl.

  2. Cryopreservation wash solution: 0.1 M Sodium Phosphate, 5% sucrose, adjust to pH 7.2 with HCl.

  3. Cryopreservation high sucrose solution: 0.1 M Sodium Phosphate, 20% sucrose, adjust to pH 7.2 with HCl.

  4. Cryopreservation embedding solution: 33% Optimal Cutting Temperature (OCT) compound diluted in cryopreservation high sucrose solution.

  5. Liquid nitrogen-cooled Isopentane.

  6. Cryostat microtome machine.

  7. Glass slides and coverslips.

  8. Wash buffer (see Note 10): PBS, 0.05% Tween 20, 50 mM Tris, 150 mM NaCl, adjust pH to 7.6 with HCl.

  9. Blocking solution: PBS, 5% goat serum, 0.05% Tween 20.

  10. Antibodies (see Note 9): Cy3-conjugated anti-human nuclear antigen (HNA), mouse anti-human CD34, mouse anti-human CD31, rabbit anti-mouse collagen type IV, rat anti-mouse CD31, Alexa488 conjugated goat anti-mouse secondary antibody, Alexa647-conjugated goat-anti rabbit secondary antibody, and Alexa647-conjugated goat-anti rat secondary antibody.

  11. Fluorescent nuclear dye: 4′,6-diamidino-2-phenylindole (DAPI).

  12. Mounting antifade medium (see Note 9).

  13. Confocal microscope with 20× objective (see Note 9).

3. Methods

3.1. Chemical Reversion of Primed hPSC to TIRN-hPSC

Because the genetic background of hPSC lines strongly contributes to interline variability, rigorously assess isogenic cultures at matched culture time points when comparing primed hPSC to TIRN-hPSC (see Note 11).

  1. Prepare all cell culture media within a sterile biological safety hood cabinet. Incubate all cells at 37 °C in a normoxic, humidified 5% CO2 incubator.

  2. Maintain primed hESC and hiPSC in feeder-free E8 medium on 6-well culture-treated dishes pre-coated with Vitronectin-XF solution (Fig. 3a; see Note 12). Replace medium daily. Passage cells every 5–6 days by either mechanical selection or non-enzymatic reagents, such as Versene or PBS-based enzyme-free cell dissociation buffer.

  3. Replace E8 medium with LIF-5i TIRN adaptation medium (see Note 13) in 4- to 5-day-old (since the last passage) hPSC cultures. The cells should be at 60–70% confluence. The next day, wash the cultures with PBS and dissociate the cells using Accutase. Transfer all of the cells at a 1:1 ratio onto a PBS-washed, gelatin- and MEF-coated plate in LIF-5i TIRN adaptation medium. Maintain culture in LIF-5i TIRN adaptation medium for one passage (4–5 days) by replacing medium daily.

  4. After 4–5 days, bulk passage the LIF-5i TIRN cells into LIF-3i TIRN conditions. Add 2 mL LIF-3i TIRN medium to each well of a gelatin- and MEF-coated PBS-washed 6-well plate. Dissociate LIF-5i TIRN adapted cells into a single cell suspension using Accutase. Resuspend the cells in LIF-5i TIRN medium and count them. Wash and transfer 0.5–1 × 106 LIF-5i TIRN-adapted single cells into each well containing LIF-3i medium. Replace LIF-3i TIRN medium daily.

  5. Passage TIRN-hESC/hiPSC in LIF-3i TIRN conditions for at least 6–7 passages using Accutase prior to use in functional or differentiation experiments (see Note 14). For each LIF-3i TIRN passage, transfer 0.2–0.5 × 106 cells per well every 3–4 days in LIF-3i TIRN medium on gelatin- and MEF-coated plates (Fig. 3a). Replace LIF-3i TIRN medium daily. Cryopreserve excess TIRN-hPSC that are not used in functional assays (see Note 15).

Fig. 3.

Fig. 3

Stepwise transition of isogenic primed hPSC to Tankyrase/PARP inhibitor-regulated naïve (TIRN)-hPSC culture conditions, and subsequent vascular directed differentiation protocol to N-VP. (a) Schematic of stepwise protocol for LIF-3i TIRN reversion of primed hPSC. (b) Schematic of APEL vascular progenitor (VP) differentiation system. AA: Activin A; B: BMP4; CHIR: CHIR99021; V: VEGF. Differentiated CD31-expressing N-VP are enriched with magnetic-activated cell sorting (MACS)

3.2. In Vitro Vascular Differentiation of N-VP from TIRN-hPSC

Efficient differentiation of TIRN-hESC/hiPSC into CD31+ CD146+ N-VP (see Note 16) has been achieved using either embryoid body-based or monolayer hemato-vascular differentiation systems [2, 6, 7]. This approach uses an improved monolayer vascular differentiation method to generate N-VP from TIRN-hPSC (summarized in Fig. 3b) and is based on a modified, previously published [2] version of the APEL medium system [29].

  1. For comparative studies (see Note 17), maintain simultaneous isogenic cultures of parental primed hPSC (in E8 medium) and TIRN-hPSC in parallel (see Note 18). One to two days after passaging TIRN-hPSC or primed hPSC (see Note 19), start the differentiation directly (without re-priming steps; see Note 20) by switching the culture media to vascular mesoderm specification medium. Maintain the culture in mesoderm specification medium for 2 days (Fig. 3b).

  2. Switch the culture medium to N-VP differentiation medium and differentiate the cells for 5–6 days (Fig. 3b). Replace N-VP differentiation medium every 2 days (see Note 21).

  3. Dissociate adherent cells differentiated for 7–8 days (i.e., following 2 days vascular mesoderm specification plus 5–6 days N-VP differentiation) using Accutase. Remove cell clumps with a 40 μm cell-strainer, count the cells, and resuspend them at a concentration of 1 × 107 cells/mL in N-VP expansion medium.

  4. Label the N-VP cells using the human CD31 microbead kit (incubate 20 μL FcR blocking reagent followed with 20 μL CD31 microbeads for 15 min at 4 °C). Enrich the differentiation cultures for CD31-expressing N-VP using LS columns and a MidiMACS separator. Prepare the columns according to the manufacturer’s instructions. After applying labeled cells to the column, wash the column 3 times with MACS wash buffer. Centrifuge the cells at 200 × g for 5 min and resuspend in N-VP expansion medium. Count the cells (see Note 22).

  5. Transfer the CD31+ N-VP cells onto fibronectin-coated plates in N-VP expansion medium at 0.1–0.2 × 106 cells per well of a 6-well plate. Grow N-VP cells to 80–90% confluency for 7–9 days prior to in vivo injections into I/R-injured murine NOG eyes, or in vitro analyses (Fig. 4a) (see Notes 23 and 24).

Fig. 4.

Fig. 4

In vitro differentiation of N-VP from TIRN-hPSC and in vivo engraftment into a humanized murine model of ischemic retinopathy. (a) Representative images of primed VP versus N-VP from a fibroblast-hiPSC after 1 passage (1 week) in EGM-2 medium post-CD31+ MACS purification, demonstrating higher human CD31 (hCD31) and Ki-67 proliferation antigen expression. (b) Humanized murine model of ischemic retinal disease for quantifying N-VP engraftment (schematic adapted from [2]). Left panel, I/R location at anterior chamber and site of human cell injections into the vitreous body. Right panel, timeline of in vivo engraftment analysis. Immunodeficient NOG mice with retinal ischemia-reperfusion (I/R) injury can be leveraged for testing human N-VP therapies by injection directly into the vitreous body. Intraocular pressure (IOP) is elevated to 120 mm Hg for 90 min in the anterior chamber, followed by vascular reperfusion, which results in retinal vasculature loss, and apoptotic death of retinal neurons ~7 days following I/R injury. Although this model does not exactly simulate the sequence of events of diabetic retinopathy, it produces the same pathology seen in all retinopathies: acellular capillaries and ischemic neuronal degeneration. (c) Representative stitched image of a whole-mount retina demonstrating survival of HNA+ N-VP cells in the superficial vascular layers of the I/R damaged retina at 7 days following intra-vitreal injection

3.3. Ocular I/R Injury and N-VP Injection into NOG Mouse Eyes

The humanized I/R ocular model utilizes transient high intraocular pressure to induce retinal ischemia-reperfusion injury (Fig. 4b), and was previously described [2, 3]. Set up an appropriate number of animals for statistically relevant analyses of retinal engraftment of human cells.

  1. Perform deep anesthesia on NOG mice by intraperitoneal (IP) injection of deep anesthesia injectable solution following procedures in line with approved local institutional and national animal use and care requirements, and in accordance with the statement on Use of Animals in Ophthalmic and Vision Research by Association for Research in Vision and Ophthalmology.

  2. Dilate pupils with 2.5% phenylephrine hydrochloride ophthalmic solution followed by 0.5% tetracaine hydrochloride eye drops as a topical anesthetic solution.

  3. Cannulate the anterior chamber of one eye under microscopic guidance (see Note 25) with a 30-gauge needle connected to a silicone infusion line providing sterile balanced salt solution. Avoid injury to the corneal endothelium, iris, and lens.

  4. Induce retinal ischemia by raising intraocular pressure (IOP) of cannulated eyes to 120 mmHg for 90 min by elevating the saline reservoir height (see Note 26).

  5. Confirm ischemia by iris whitening and loss of retinal red reflex. Maintain anesthesia with two doses of intramuscular ketamine solution for up to 90 min. Withdraw the needle to normalize IOP, and confirm reperfusion of retinal blood flow by evaluating reappearance of the red reflex.

  6. Leave the contralateral eye of each animal unmanipulated to serve as a non-ischemic negative control.

  7. Apply ophthalmic topical antibiotic ointment.

  8. Two days after I/R injury, inject expanded human N-VP cells into the vitreous body of I/R-treated eye. Use 50,000 cells in a maximum of 2 μL per eye in a 1:1 sterile mixture of EGM-2/PBS using a digital pico-injector.

3.4. Harvest of NOG Mouse Retinae

Human N-VP cell engraftment in the NOG mouse retinae can be detected directly with anti-human nuclear antigen (HNA) immunohistochemistry, along with murine vascular marker co-localization using anti-murine CD31 and collagen IV antibodies.

  1. Euthanize animals by cervical dislocation (see Note 27) for retinal harvests according to approved local institutional and national animal use and care protocols (see Note 28). Prepare the appropriate number of retinae for downstream analyses of HNA-positive cell quantitation in whole-mount retinae and human vascular cell quantitation by cross-section evaluations at desired post-treatment intervals (e.g., 1, 4, 8, 12 weeks) following human N-VP (or primed VP) injections (i.e., 2 days post-I/R injury).

  2. Enucleate eyes, remove cornea and lens, and carefully separate the retinae from the choroid and sclera with a blunt cyclodialysis spatula. Fix retinae in fixative solution (Subheading 2.3) overnight at 4 °C. Transfer tissues to sterile PBS before processing eyes for whole-mount retinae or cross-sections.

3.5. Preparation and Imaging of Flat Whole-Mounted NOG Mouse Retinae for Human Engraftment Analysis by HNA Expression

  1. For whole-mount retinae, permeabilize cells by incubation with permeabilization solution in an Eppendorf tube for 15 min at 4 °C.

  2. Thoroughly wash the tissues with TBS. Block free-floating retinae with blocking solution (Subheading 2.3) for 6–8 h. Incubate the tissues overnight at 4 °C in primary antibody solutions diluted in blocking solution: rabbit anti-mouse Collagen IV (1:100) and/or rat anti-mouse CD31 (1:50) to label basement membrane and endothelial cells of blood vessels, respectively.

  3. On the next day, wash retinae with wash buffer, and incubate the tissues with secondary antibodies for 6 h at 4 °C. A goat anti-rabbit Cy3-conjugated secondary antibody can be used to detect the collagen IV primary antibody, and a goat anti-rat Alexafluor-647-conjugated secondary antibody can be used to detect the anti-CD31 primary antibody. Human cells can be detected using directly Cy3-conjugated anti-HNA (1:100). Counterstain nuclei using a fluorescent nuclear dye.

  4. After removing excess antibody and nuclear dye with wash buffer, pie-cut retinae and place flattened retinae on glass slides (see Note 29). Mount the slides with a coverslip (see Note 30). Image flat-mount retinae with a confocal microscope (see Note 31). For each individual eye, the entire retina can be imaged by tile-scanning and stitching (7 × 7 frames, 10% overlapping).

  5. Quantitate human (HNA+) cell vascular engraftment in stitched photomicrographs of whole-mount murine retinae using a microscopy analysis software (Fig. 4c) (see Note 32). First, conservatively delineate the whole retina by creating a region of interest (ROI) using the nuclear dye channel (see Note 33). Exclude any background in the Cy3 channel at the edges of the retina tissue by adjusting the ROI (see Note 34). Process the Cy3 HNA channel with the “smooth” function and create a mask by thresholding. Exclude non-specific areas outside the ROI. Quantitate the number of HNA+ nuclei in the Cy3 channel using the “analyze particle” plugin (if using ImageJ) by using standardized sequential corrections (see Note 35). Identify HNA+ nuclei as particle objects by automated counting using fixed size and circularity parameters.

3.6. Preparation and Imaging of NOG Mouse Neural Retinal Cross-Sections for Quantitation of Human Vascular Engraftment

Human CD34+ and CD31+ cell engraftment can be detected and quantified within the defined neural layers of the murine retina by analyzing retinal cross-sections with the following methods (Fig. 5).

Fig. 5.

Fig. 5

Method of human CD31+ engraftment quantitation in the vasculature of the neural layers (ILM, GCL, IPL, INL, OPL, ONL) of ischemia-injured mouse retinae. Shown is a representative example of ROI defining the neural retinal layers evaluated for human chimeric vascularization with murine blood vessels. A method of regional separation of neural retinal layers was employed for quantification of human CD31+ (hCD31) or alternatively CD34+ cells via automated counts (Analyze particles function) using the Fiji distribution of ImageJ software

  1. Dissect the anterior eye (cornea/iris) free by a circumferential cut at the limbus. Fix eyecups in fixative solution (Subheading 2.4) for 1–2 h at room temperature. Wash 3 times for 10 min in cryopreservation wash solution. Prepare for cryopreservation by immersion in gradients of sucrose using successive 2:1, 1:1, 1:2 mix ratios of cryopreservation wash solution and cryopreservation high sucrose solution [30]. Keep tissues in cryopreservation high sucrose solution overnight at 4 °C. The next day, hemisect eyes through the optic nerve, and embed the two halves in cryopreservation embedding solution. Freeze in isopentane cooled with liquid nitrogen. Serially cryosect retinal hemi-sections (8 μm thickness) using a cryostat microtome. Transfer sections onto glass slides and store at −80 °C.

  2. Label equally interspaced microsections with either anti-human CD34 or CD31 antibodies (see Note 36). Let the cryopreserved slides stabilize at room temperature and wash 3 times for 5 min at room temperature in wash buffer (Subheading 2.4). Incubate retinal sections for 1 h at room temperature in blocking solution (Subheading 2.4). Remove excess blocking solution, and without washing, incubate the retinal sections overnight at 4 °C with either mouse anti-human CD34 (1:50) or mouse anti-human CD31 (1:50) antibodies that are diluted in blocking solution. Wash tissues 3 times in wash buffer. Incubate tissue with Alexa 488-conjugated secondary goat anti-mouse antibody (1:200) for 1 h at room temperature (see Note 37). Wash 3 times in wash buffer. Label sections with either rabbit anti-mouse collagen type IV (1:200) or rat anti-mouse CD31 (1:50) diluted in blocking solution for 1 h at room temperature. Wash 3 times in wash buffer. Incubate with Alexa 647-conjugated goat-anti rabbit (1:200) or goat anti-rat secondary antibodies (see Note 38) diluted in blocking solution for 1 h at room temperature. Counterstain nuclei with DAPI.

  3. Mount retinal sections with a glass coverslip in mounting antifade medium and cure overnight in the dark. Acquire images using a 20× objective with a confocal microscope.

  4. Quantitate the number of human CD34 or CD31 blood vessels within the defined neural layers of the murine retina using microscopic image analysis software (Fig. 5) (see Note 32). Create three ROIs using the DAPI channel as a template, and delineate the ganglion cell layer (GCL), inner nuclear layer (INL), and outer nuclear layer (ONL). The other regions are defined as intercalated around and between these three DAPI-defined ROIs. Switch to the Alexa 488 (either human CD34 or CD31) channel and process the image for quantitation using a fixed sequential series of defined parameters. We recommend applying the “smooth”, “despeckle”, and “filter (median)” functions (see Note 39). Proceed with thresholding the images (pixel values >50). Count the number of thresholded objects within and between ROIs (see Note 40). The number of human CD34+ or CD31+ blood vessels can be automatically captured and enumerated using the “Analyze particles” plugin in ImageJ (see Note 41). Cross-validate each automatic count by manually counting the objects within the thresholded images and exclude any duplicate objects (see Note 42). To increase the precision of human CD34 or CD31 expression quantitation, check the Alexa647 channel and score only the regions where the Alexa488+ signal is detected at chimeric human-murine blood vessels that also express either murine collagen IV (mColIV) or murine CD31 (mCD31).

Acknowledgments

This work was supported by grants from the NIH/NEI (R01EY032113; ETZ); R01EY023962; ETZ) R01EY016151; GL), EY01765; Wilmer Eye Institute), NIH/NICHD (R01HD082098; ETZ), The Maryland Stem Cell Research Fund (2020-MSCRFD-5374; ETZ), and The Lisa Dean Moseley Foundation (ETZ).

4 Notes

1.

Prepare E8 medium as reported [31] or purchase commercially. For cost savings, the FGF2 concentration can be reduced to 50 ng/mL without impacting the pluripotent phenotype of hESC and hiPSC lines. To preserve FGF2 stability, we recommend avoiding prolonged pre-warming of E8 medium.

2.

Minimizing the amount of dimethylsulfoxide (DMSO) added to the TIRN medium is recommended by preparing stocks of the small molecules (CHIR99021, PD0325901, and XAV939) at high concentrations (e.g., 100 mM). Freshly prepared TIRN medium should be prewarmed before being sterile filtered to ensure small molecule solubility.

3.

MEF feeders are commercially available, or can be prepared from E13.5 CF1 or hybrid CF1 × DR4 mouse embryos using published protocols [32]. We routinely cryopreserve low passage MEF cultures pre- (less than 2 passages) or post- (up to passage 5) irradiation (5000 rad). Cryopreserved MEF are stored in liquid nitrogen, and freshly thawed 24–48 h before TIRN-hESC and -hiPSC passaging.

4.

N-VP (and primed VP) differentiation has been optimized using the commercially available STEMdiff APEL 2-LI medium (StemCell Technologies) for hemato-vascular mesodermal differentiation.

5.

MACS-enriched CD31+ N-VP co-express CD146. CD31+CD146+ N-VP populations are further expanded for several passages in EGM-2 medium prior to in vitro or in vivo analyses (Fig. 1).

6.

NOG animals are immunodeficient and all food, water, bedding, and cages should be sterilized. An individually ventilated cage system is recommended. Aged animals may exhibit variable vascular remodeling. Animal surgical procedures should be performed in accordance with protocols approved by national and local institutional regulations, such as the Institutional Animal Care and Use Committees (IACUC).

7.

Use an anterior chamber cannulation microscope.

8.

We use a commercially available 4% stock solution of paraformaldehyde. Preparation of fresh paraformaldehyde solution from powder is not advisable.

9.

We use Cy3-conjugated anti-HNA antibody from Millipore (MAB1281C3), rabbit anti-mouse Collagen IV from Millipore (AB756P), rat anti-mouse CD31 from Biosciences (550274), mouse anti-human CD34 from BD Biosciences (clone My10, #347660), mouse anti-human CD31 from Dako (M0823), rabbit anti-mouse collagen type IV from Millipore (AB756P; 1:100), and rat anti-mouse CD31 from BD Biosciences. (550274; 1:50). We recommend using fluorochrome-conjugated F(ab’)2 fragment secondary antibodies that are highly cross-adsorbed. Anti-fade mounting medium is purchased from ThermoFisher Scientific (Prolong Gold). We routinely analyze fluorescent images using a LSM510 Meta confocal microscope with a 20× objective.

10.

We routinely purchase Wash Buffer 10× from Agilent Dako and dilute to a 1× working concentration.

11.

Some naïve hPSC culture systems generate populations with aberrant genomic and epigenetic configurations, as well as defective differentiation capacity [20]. Naïve reversion methods should be assayed with multiple independent genetic backgrounds, in a manner sufficient to validate biological reproducibility and exclude developmentally irrelevant “pseudo-pluripotent” states. Accordingly, studies of human naïve culture systems have focused on assaying molecular pluripotency of hPSC at (1) the epigenetic level (e.g., histone marks by ChIP sequencing or ChIP-PCR, global and allele-specific CpG methylation analysis, POU5F1 (OCT4) enhancer usage, activity at DNase I hypersensitivity regulatory elements), (2) transcriptomic level (RNA-sequencing, expression microarrays, and quantitative RT-PCR), protein expression (e.g., FACS, immunofluorescent microscopy, and Western blotting) and (3) metabolic studies (e.g., glycolysis, oxidative phosphorylation, and nicotinamide metabolism).

12.

All investigations with human stem cells should be derived from appropriately consented human donor tissues that have undergone rigorous review by Institutional Stem Cell Research Oversight Committees and Institutional Review Board Committees, where applicable. All primed hPSC lines should be validated for normal karyotypes before TIRN reversion. Primed hPSC lines should be reverted at the lowest possible available passage. Naïve reversion of high-passage primed hPSC lines (e.g., passage number > 40) should be avoided, as such cultures often already harbor culture-associated genomic aberrations that can negatively impact efficient stable TIRN reversion.

13.

Primed hPSC should be adapted for one passage in LIF-5i TIRN adaptation medium prior to switching to LIF-3i TIRN medium. This step facilitates the initial clonal re-plating efficiencies of primed hPSC lines across genetic backgrounds [6, 7]. The use of anti-apoptotic reagents (e.g., 5–10 μM Y-27632) is not generally required with the LIF-5i TIRN adaptation method. The transition from primed cells to TIRN culture systems is accompanied by distinct changes in hESC/hiPSC colony morphology. For example, primed hPSC expand as flat, wide monolayer colonies from small cell clumps (on MEF or in feeder-free conditions), but poorly as single cells [6, 7]. In contrast, reversion of primed hPSC lines in TIRN medium promotes the rapid growth and expansion of small, tightly packed, dome-shaped colonies that arise clonally from single cells. However, these morphological changes are reversible, and TIRN-reverted dome-shaped colonies can spontaneously transition back to a conventional monolayer morphology if TIRN medium is withdrawn and cells are re-cultured in primed hESC medium supplemented with FGF2. Additionally, please note that the growth of TIRN-hPSC at high densities results in spontaneous reacquisition of the flat, conventional morphology. Finally, although this protocol begins with feeder-free primed hPSC cultured in E8 medium, we have also published detailed protocols for TIRN reversion of MEF-dependent primed hESC and hiPSC [6, 7].

14.

The LIF-3i TIRN method has been successfully applied to a broad repertoire of >30 independent normal and diabetic primed hPSC lines. LIF-3i TIRN-reverted hESC and hiPSC adopted a naïve-epiblast-like state for >20 passages without evidence of chromosomal or epigenomic abnormalities, including at imprinted genomic loci [2, 6, 7]. High confluency/cell density of TIRN-hPSC cultures should be avoided as this decreases clonal re-plating efficiency, and promotes spontaneous differentiation.

15.

Excess TIRN-hESC and hiPSC can be frozen in TIRN cryopreservation medium. We recommend aliquoting 1–1.5 × 106 TIRN-hPSC (that have been naïve-reverted for at least 3 passages after TIRN adaptation) per cryovial for slow freezing. To thaw cryopreserved cells, add 5 μM Y-27632 to LIF-3i TIRN medium overnight post-thaw. Freshly cryopreserved TIRN-hPSC should be expanded for at least 3 passages in TIRN medium before use in functional assays.

16.

The TIRN method is the only naïve reversion method reported thus far that improves the functional pluripotency of a broad repertoire of hPSC lines by decreasing their lineage-primed gene expression and dramatically improving their multipotent differentiation potency [2, 6, 7]. TIRN reversion also diminishes the inherent interline variability of differentiation of lineage-primed, conventional hPSC lines.

17.

For functional comparisons of primed versus TIRN-hPSC, set up parallel cultures from the same isogenic hPSC line at an equivalent passage number (Fig. 3b) [2, 6, 7]. To eliminate experimental bias, maintain primed/naïve sibling isogenic hPSC cultures in parallel in their respective media (e.g., E8 medium versus TIRN medium), and simultaneously differentiate the cells in parallel using identical differentiation protocols and materials. For isogenic primed versus naïve hPSC comparisons, adjust and optimize initial plating densities for each individual differentiation assay. Detailed protocols for neural progenitor, definitive endoderm and hemato-endothelial directed differentiations of TIRN-hPSC are provided elsewhere [2, 6, 7].

18.

Record the number of passages hPSC have been cultured in both TIRN or primed media. We recommend starting all functional studies with fresh TIRN reversions of low-passage primed hiPSC lines. The use of hESC or hiPSC that have undergone greater than ten passages in TIRN medium is not recommended for functional studies; rapidly proliferating TIRN-hPSC cultures may potentially select karyotypically abnormal clones.

19.

TIRN-hPSC have a more robust proliferative and differentiation capacity in directed differentiation assays than primed hPSC. TIRN-hPSC also yield higher numbers of differentiated cells than conventional hPSC.

20.

TIRN-hPSC should be directly utilized in established directed differentiation protocols without additional cell culture manipulations. For example, “re-priming” (i.e., converting N-hPSC back to primed conditions prior to their use in directed differentiation assays) is not necessary and is not recommended with the TIRN method. To control for impacts of assay and interline variability of individual hPSC lines, cross-validate lineage-specific differentiation potencies by employing independent differentiation protocols with at least three hPSC lines derived from independent genetic backgrounds.

21.

After 7–8 days, differentiated N-VP (or primed VP) cells reach full confluence and cultures should include large cell areas that adopt typical endothelial cobblestone monolayer morphology.

22.

CD31+ N-VP co-express CD146 post-MACS enrichment. We routinely perform post-MACS enrichment analysis using flow cytometry to measure cell purity using APC-conjugated mouse anti-CD31 and PE-conjugated mouse anti-CD146 (Clone P1H12; BD Biosciences) staining.

23.

We routinely perform in vitro functional analyses of CD31+CD146+ N-VP to validate functionality prior to in vivo transplantation. These analyses include vascular marker expression (e.g., flow cytometry, Western blot, immunofluorescence, RT-PCR, RNA-sequencing), vascular functional assays (e.g., Matrigel tube forming assays, acetylated Dil-LDL, and UEA1 lectin uptake assays), cell cycle assays, senescence assays, and DNA damage assays [2, 3].

24.

TIRN-hPSC routinely demonstrated more robust vascular differentiation in vitro than isogenic primed hPSC counterparts. For example, directed TIRN-hPSC vascular differentiation results in more rapid kinetics of multipotent N-VP expression (CD31+ CD146+), pericytic progenitors (CD34+ CD140b+), endothelial progenitors (CD144+ CD105+ CD31+), and other mesodermal vascular populations (KDR+ CD144+) than from their isogenic primed hPSC counterparts. TIRN-hPSC cultures also generated higher frequencies of cobblestone endothelial CD31+ cell monolayers possessing higher proliferative (Ki-67+) rates, and acetylated Dil-LDL-binding endothelial functionality [2, 3].

25.

Surgical procedures are performed with a Zeiss OPMI VISU 200 surgical microscope within a HEPA filtered sterile surgical suite. Cannulation of the anterior chamber is performed with a 30-gauge needle that infuses sterile balanced salt solution to elevate IOP.

26.

The elevation of the reservoir height is not standardized between IR injury model studies and should be validated [33, 34]. Other protocols have reported IOP elevation during ischemia induction ranging from 60 to 120 mmHg.

27.

Methods of euthanasia influence retinal physiology [35].

28.

The time required for performing I/R ocular injury model on large numbers of mice may require staggering the experimental design. Efforts should be made for comparative studies to match cell populations and animals for each time point. Control non-treated eyes can best be obtained by using the contralateral eye of the same mouse.

29.

Orientate by placing the outer layer retina on slide and the superficial layer facing toward the top.

30.

Antifade mounting reagent can be used, or alternatively retinae can be placed in wash buffer if imaged immediately.

31.

We routinely acquire images with ZEN software using a 10× objective and a LSM510 Meta confocal microscope.

32.

We recommend using the Fiji distribution of ImageJ [36].

33.

We routinely use the “magic wand” function in ImageJ software.

34.

ROI created using the magical wand function can be easily adjusted. Pie-cut retinae may show limited background in fluorescent channels at the edges of the tissue. These non-specific labeled cells should be excluded from the analysis. If using Image J, eliminate noise in the DAPI channel by applying the “Minimum” filter, create a new ROI with the magical wand and use the function “Clear outside” to eliminate all signal outside the retina area.

35.

Our ImageJ software workflow settings are limited to “smooth”, “threshold” (e.g., interMode 50), “despeckle”, and “watershed”.

36.

Perform negative immunostaining controls for each individual experiment to determine antibody specificity by replacing primary antibodies with mouse, rat, and rabbit nonimmune IgG at the corresponding antibody concentration to verify absence of unspecific antibody binding.

37.

Other fluorochromes may also be used (e.g., Alexa 555 or Cy3).

38.

We recommend using conjugated F(ab’)2 fragments or secondary antibodies that are highly cross-adsorbed against IgG from other species.

39.

Speckles and non-specific background (<10 pixels) should be excluded.

40.

Depending on the number of Alexa 488-positive objects per cross-section, objects may be individually counted.

41.

The layers of the whole retina should fill the microscopic field when centered using a 20× objective. The surface area of analyzed cross-sections using a 20× objective and a LSM510 Meta confocal microscope is 450 μm2. We recommend setting up the ImageJ module “Analyze particles” to select objects with a surface area >5 μm2.

42.

Caution: because blood vessels may be longitudinally cross-sectioned, two or more objects may belong to the same vessel. We recommend deducting closely associated objects.

References

  • 1.Linask KK, Han M, Bravo-Valenzuela NJ (2014) Changes in vitelline and utero-placental hemodynamics: implications for cardiovascular development. Front Physiol 5:390. 10.3389/fphys.2014.00390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Park TS, Zimmerlin L, Evans-Moses R et al. (2020) Vascular progenitors generated from tankyrase inhibitor-regulated naive diabetic human iPSC potentiate efficient revascularization of ischemic retina. Nat Commun 11(1):1195. 10.1038/s41467-020-14764-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Park TS, Bhutto I, Zimmerlin L et al. (2014) Vascular progenitors from cord blood-derived induced pluripotent stem cells possess augmented capacity for regenerating ischemic retinal vasculature. Circulation 129(3):359–372. 10.1161/CIRCULATI0NAHA.113.003000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Park TS, Zimmerlin L, Zambidis ET (2013) Efficient and simultaneous generation of hematopoietic and vascular progenitors from human induced pluripotent stem cells. Cytometry A 83(1):114–126. 10.1002/cyto.a.22090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zambidis ET, Sinka L, Tavian M et al. (2007) Emergence of human angiohematopoietic cells in normal development and from cultured embryonic stem cells. Ann N Y Acad Sci 1106:223–232. 10.1196/annals.1392.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zimmerlin L, Park TS, Huo JS et al. (2016) Tankyrase inhibition promotes a stable human naive pluripotent state with improved functionality. Development 143(23):4368–4380. 10.1242/dev.138982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Park TS, Zimmerlin L, Evans-Moses R et al. (2018) Chemical reversion of conventional human pluripotent stem cells to a naive-like state with improved multilineage differentiation potency. J Vis Exp (136):e57921. 10.3791/57921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zambidis ET, Peault B, Park TS et al. (2005) Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development. Blood 106(3):860–870. 10.1182/blood-2004-11-4522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Majesky MW, Dong XR, Regan JN et al. (2011) Vascular smooth muscle progenitor cells: building and repairing blood vessels. Circ Res 108(3):365–377. 10.1161/CIRCRESAHA.110.223800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zimmerlin L, Park T, Donnenberg V et al. (2014) Pericytes: a ubiquitous source of multipotent adult tissue stem cells. In: Shiffman MA, Di Giuseppe A, Bassetto F (eds) Stem cells in aesthetic procedures. Springer, Berlin, pp 135–148. 10.1007/978-3-642-45207-9_9 [DOI] [Google Scholar]
  • 11.Vodyanik MA, Yu J, Zhang X et al. (2010) A mesoderm-derived precursor for mesenchymal stem and endothelial cells. Cell Stem Cell 7(6):718–729. 10.1016/j.stem.2010.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Brunner S, Hoellerl F, Schmid-Kubista KE et al. (2011) Circulating angiopoietic cells and diabetic retinopathy in type 2 diabetes mellitus, with or without macrovascular disease. Invest Ophthalmol Vis Sci 52(7):4655–4662. 10.1167/iovs.10-6520 [DOI] [PubMed] [Google Scholar]
  • 13.Caballero S, Sengupta N, Afzal A et al. (2007) Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes 56(4):960–967. 10.2337/db06-1254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Jarajapu YP, Grant MB (2010) The promise of cell-based therapies for diabetic complications: challenges and solutions. Circ Res 106(5):854–869. 10.1161/CIRCRESAHA.109.213140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Tan K, Lessieur E, Cutler A et al. (2010) Impaired function of circulating CD34(+) CD45(−) cells in patients with proliferative diabetic retinopathy. Exp Eye Res 91(2):229–237. 10.1016/j.exer.2010.05.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Li Calzi S, Neu MB, Shaw LC et al. (2010) Endothelial progenitor dysfunction in the pathogenesis of diabetic retinopathy: treatment concept to correct diabetes-associated deficits. EPMA J 1(1):88–100. 10.1007/s13167-010-0011-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dar A, Domev H, Ben-Yosef O et al. (2011) Multipotent vasculogenic pericytes from human pluripotent stem cells promote recovery of murine ischemic limb. Circulation 125(1):87–99. 10.1161/CIRCULATIONAHA.111.048264 [DOI] [PubMed] [Google Scholar]
  • 18.Kyttala A, Moraghebi R, Valensisi C et al. (2016) Genetic variability overrides the impact of parental cell type and determines iPSC differentiation potential. Stem Cell Reports 6(2):200–212. 10.1016/j.stemcr.2015.12.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Feng Q, Lu SJ, Klimanskaya I et al. (2010) Hemangioblastic derivatives from human induced pluripotent stem cells exhibit limited expansion and early senescence. Stem Cells 28(4):704–712. 10.1002/stem.321 [DOI] [PubMed] [Google Scholar]
  • 20.Zimmerlin L, Park TS, Zambidis ET (2017) Capturing human naive pluripotency in the embryo and in the dish. Stem Cells Dev 26(16):1141–1161. 10.1089/scd.2017.0055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Khullar M, Cheema BS, Raut SK (2017) Emerging evidence of epigenetic modifications in vascular complication of diabetes. Front Endocrinol (Lausanne) 8:237. 10.3389/fendo.2017.00237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Manzar GS, Kim EM, Zavazava N (2017) Demethylation of induced pluripotent stem cells from type 1 diabetic patients enhances differentiation into functional pancreatic beta cells. J Biol Chem 292(34):14066–14079. 10.1074/jbc.M117.784280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yang J, Ryan DJ, Wang W et al. (2017) Establishment of mouse expanded potential stem cells. Nature 550(7676):393–397. 10.1038/nature24052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gao X, Nowak-Imialek M, Chen X et al. (2019) Establishment of porcine and human expanded potential stem cells. Nat Cell Biol 21(6):687–699. 10.1038/s41556-019-0333-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mandai M, Kurimoto Y, Takahashi M (2017) Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med 377(8):792–793. 10.1056/NEJMc1706274 [DOI] [PubMed] [Google Scholar]
  • 26.da Cruz L, Fynes K, Georgiadis O et al. (2018) Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol 36(4):328–337. 10.1038/nbt.4114 [DOI] [PubMed] [Google Scholar]
  • 27.Stacey GN, Crook JM, Hei D et al. (2013) Banking human induced pluripotent stem cells: lessons learned from embryonic stem cells? Cell Stem Cell 13(4):385–388. 10.1016/j.stem.2013.09.007 [DOI] [PubMed] [Google Scholar]
  • 28.Taylor CJ, Bolton EM, Pocock S et al. (2005) Banking on human embryonic stem cells: estimating the number of donor cell lines needed for HLA matching. Lancet 366(9502):2019–2025. 10.1016/S0140-6736(05)67813-0 [DOI] [PubMed] [Google Scholar]
  • 29.Ng ES, Davis R, Stanley EG et al. (2008) A protocol describing the use of a recombinant protein-based, animal product-free medium (APEL) for human embryonic stem cell differentiation as spin embryoid bodies. Nat Protoc 3(5):768–776. 10.1038/nprot.2008.42 [DOI] [PubMed] [Google Scholar]
  • 30.McLeod DS, Grebe R, Bhutto I et al. (2009) Relationship between RPE and choriocapillaris in age-related macular degeneration. Invest Ophthalmol Vis Sci 50(10):4982–4991. 10.1167/iovs.09-3639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chen G, Gulbranson DR, Hou Z et al. (2011) Chemically defined conditions for human iPSC derivation and culture. Nat Methods 8(5): 424–429. 10.1038/nmeth.1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Jozefczuk J, Drews K, Adjaye J (2012) Preparation of mouse embryonic fibroblast cells suitable for culturing human embryonic and induced pluripotent stem cells. J Vis Exp (64):3854. 10.3791/3854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Inafuku S, Klokman G, Connor KM (2018) The alternative complement system mediates cell death in retinal ischemia reperfusion injury. Front Mol Neurosci 11:278. 10.3389/fnmol.2018.00278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zheng L, Gong B, Hatala DA et al. (2007) Retinal ischemia and reperfusion causes capillary degeneration: similarities to diabetes. Invest Ophthalmol Vis Sci 48(1):361–367. 10.1167/iovs.06-0510 [DOI] [PubMed] [Google Scholar]
  • 35.Hwang CK, Iuvone PM (2013) Technical brief: a comparison of two methods of euthanasia on retinal dopamine levels. Mol Vis 19:1122–1124 [PMC free article] [PubMed] [Google Scholar]
  • 36.Schindelin J, Arganda-Carreras I, Frise E et al. (2012) Fiji: an open-source platform for biological-image analysis. Nat Methods 9(7):676–682. 10.1038/nmeth.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES