- Review
- Open access
- Published:
Ion channels research in hPSC-RPE cells: bridging benchwork to clinical applications
Journal of Translational Medicine volume 22, Article number: 1073 (2024)
Abstract
Ion channels in retinal pigment epithelial (RPE) cells are crucial for retinal health and vision functions. Defects in such channels are intricately associated with the development of various retinopathies that cause blindness. Human pluripotent stem cells (hPSC)-derived RPE cells, including those from human-induced pluripotent stem cells (hiPSC) and human embryonic stem cells (hESC), have been used as in vitro models for investigating pathogenic mechanisms and screening potential therapeutic strategies for retinopathies. Therefore, the cellular status of hPSC-RPE cells, including maturity and physiologic functions, have been widely explored. Particularly, research on ion channels in hPSC-RPE cells can lead to the development of more stable models upon which robust investigations and clinical safety assessments can be performed. Moreover, the use of patient-specific hiPSC-RPE cells has significantly accelerated the clinical translation of gene therapy for retinal channelopathies, such as bestrophinopathies. This review consolidates current research on ion channels in hPSC-RPE cells, specifically Kir7.1, Bestrophin-1, CLC-2, and CaV1.3, providing a foundation for future research.
Introduction
Retinal pigment epithelial (RPE) cells constitute a polarized monolayer between photoreceptors and Bruch membrane. The RPE cells participate in the maintenance of normal visual function through establishing the outer blood-retinal barrier, phagocytosing shed photoreceptor outer segments (POS), regulating hydro-ionic homeostasis within the subretinal space, supporting the retinoid cycle, secreting growth factors, and absorbing scattered light [1, 2]. Ion channels are crucial for maintaining these functions. For instance, Kir7.1 (potassium channel), Bestrophin-1 (chloride channel), CaV1.3 (calcium channel), and NaV1.4 (sodium channel) are all involved in the regulation of POS phagocytosis [3,4,5,6]. Disruption of certain ion channels in RPE cells can lead to various retinopathies, including bestrophinopathies [7], retinitis pigmentosa (RP) [8], Leber congenital amaurosis (LCA) [9], and age-related macular degeneration (AMD) [10]. Thus, the role of ion channels in RPE cells warrants comprehensive evaluation.
Over decades, various effective methods have been established for generating RPE cells from human embryoid stem cells (hESC) and human induced pluripotent stem cells (hiPSC) [11,12,13]. Notably, hiPSC- and hESC-derived RPE cells differ in their genetic and ethical aspects. hiPSC-RPE cells have a genetic makeup identical to the donor’s somatic cells, and mitigate ethical concerns surrounding the utilization of embryonic cells. However, individualized hiPSC-RPE cells are costly and require longer preparation time. Moreover, the process of reprogramming somatic cells into hiPSC may introduce novel pathogenetic mutations or epigenetic modifications that could potentially affect their functionality and stability [14]. Despite these differences, both hiPSC- and hESC-RPE cells are classified as human pluripotent stem cell-derived RPE (hPSC-RPE) cells due to their nearly indistinguishable morphological and functional characteristics. These cells highly express key RPE-specific markers, including RPE65, CRALBP, MITF, Bestrophin-1, and MERTK, and effectively recapitulate critical RPE functions, such as phagocytosis of POS, secretion of VEGF/PEDF, and participation in the retinoid cycle [15, 16]. Electrophysiological studies have shown that hPSC-RPE cells possess membrane potentials and ionic currents comparable to those observed in freshly isolated RPE cells, indicating a degree of functional maturity that closely resembles primary human RPE cells [1, 2]. Consequently, hPSC-RPE cells have become invaluable tools for modeling retinal diseases, investigating pathogenetic mechanisms, and advancing therapeutic approaches such as gene therapy, cell transplantation, and high-throughput drug screening [17,18,19].
Research on ion channels in hPSC-RPE cells has significantly advanced the clinical translation of therapies for retinopathies. It has deepened our understanding of the pathogenic mechanisms and therapeutic treatments for channelopathies, including LCA16, CLCN2-IRD, and bestrophinopathies [7, 20, 21]. Additionally, it has surpported the development of hPSC-RPE cells transplantation therapy for patients with other retinopathies, such as AMD and Stargardt disease (STGD) [22, 23]. Although research on ion channels in hPSC-RPE cells has surged in recent years, no comprehensive review has been published. This review consolidates and critically evaluates existing studies on ion channels in hPSC-RPE cells, highlighting their potential clinical applications.
Potassium channels
Potassium (K+) channels are one of the most common ion channel families in RPE cells. In this study, we explored the spectrum of findings from potassium channel research in hPSC-RPE cells, especially inwardly rectifying potassium channels (Kir) and voltage-gated potassium channels (KV). The KV family has 40 members across 12 subfamilies and is crucial for generating membrane potential and regulating cell volume by facilitating potassium efflux in excitable cells [24]. KV also induces electrical excitability in RPE cells [25, 26]. Recent studies have investigated the expression of various KV members, such as KV1.3 (KCNA3), KV1.4 (KCNA4), KV4.2 (KCND2), and KV7.1 to KV7.5 (KCNQ1-KCNQ5) in hESC-RPE cells (Fig. 1 and Suppl. Table 1) [27]. These channels produce diverse potassium currents, including delayed rectifier currents inhibited by Agitoxin-2 (targeting KV1.3) and the sustained M-currents blocked by Linopirdine (targeting KV) [27]. Additionally, fast-activating currents generated by KV1.4 and KV4.2 resemble the transient A-type currents reported in native human RPE cells [27, 28]. The large-conductance calcium-activated potassium channels (maxi-K) can be classified as voltage-activated K+ channels. A recent study showed that maxi-K is expressed in the apical and basolateral membrane of hiPSC-RPE cells [29]. Blocking the maxi-K significantly reduces the secretion of basolateral VEGF and apical PEDF; however, it does not disrupt the POS internalization in the cells [29].
Potassium and chloride channels in hPSC-RPE cells. The subcellular localization of chloride channels (Bestrophin-1, CLC-2) and potassium channels (Kir4.1, Kir7.1, KV1.3, KV1.4, KV4.2, KV7.1-KV7.5, maxi-K) in hPSC-RPE cells. Although CLIC4 is detectable in hPSC-RPE cells, its subcellular localization has not yet been described; hence, its localization is inferred based on observations from ARPE-19 cells. Abbreviations: sER, smooth endoplasmic reticulum; rER, rough endoplasmic reticulum; GC, Golgi complex; Mit, mitochondrion
Kir channels are essential for maintaining potassium homeostasis within cells. Although eight Kir channels (Kir1.1, Kir2.1, Kir2.2, Kir3.1, Kir3.4, Kir4.2, Kir6.1, and Kir7.1) have been identified in human RPE cells [30, 31], only Kir7.1 has been extensively studied in hPSC-RPE cells. The research progress of Kir7.1 in hPSC-RPE cells is detailed below. The other seven Kir channels show significantly lower transcript levels than Kir7.1 [30]; however, their potential contributions to RPE cell function should not be underestimated. For instance, Kir4.1 is located in the apical membrane, particularly enriched in the microvilli of both hESC- and hiPSC-RPE cells, although its role remains poor understood [27, 29]. In summary, aside from Kir7.1, research on other potassium channels in hPSC-RPE cells is still in the preliminary stages, hilighting the need for more in-depth studies to elucidate their roles and functions.
Kir7.1
Function and unique characteristics of Kir7.1
Kir7.1, encoded by the KCNJ13 on chromosome 2q37, functions as an inwardly rectifying potassium channel. Its activity is modulated by several factors, including ATP, phosphatidylinositol 4,5-bisphosphate (PIP2), G proteins, sodium ions, and protons [32, 33]. Kir7.1 is a highly divergent member of the Kir channel family, sharing approximately 50% protein sequence homology with its closest relatives, Kir4.1 and Kir4.2 [34]. This distinct amino acid sequence may confer specialized functions to Kir7.1 in RPE cells, including the maintenance of potassium ion homeostasis and the regulation of POS phagocytosis [5, 35, 36]. Kir7.1 is highly expressed on the apical membrane of native human RPE cells [27, 37] and hiPSC- or hESC-derived RPE cells [5, 27, 29, 38]. Intriguingly, Kir7.1 has also been identified on the basolateral membrane in hESC-RPE cells [27], likely due to differences in culture conditions and cultivation time. These findings suggest that Kir7.1 could serve as a valuable biomarker to assess the polarity and maturation status of hPSC-RPE cells for clinical transplantation.
Advances in KCNJ13-IRD pathogenesis and treatment strategies
Mutations in KCNJ13 lead to two types of channelopathies, including Snowflake vitreoretinal degeneration (SVD, MIM#193230) [39] and Leber congenital amaurosis 16 (LCA16, MIM#614186) [20, 40]. SVD is a very rare autosomal dominant inherited retinal disease (< 1/1000000), characterized by early-onset cataracts, vitreous degeneration, and peripheral retinal abnormalities (minute crystalline-like deposits called snowflakes) [34]. LCA16 is a congenital autosomal recessive retinal degeneration disease characterized by significant disruption of both RPE and photoreceptors, leading to severe visual impairment in patients from the first year of life [20, 40]. Both channelopathies underscore the indispensable role of Kir7.1 in maintaining normal retinal homeostasis and function. Although homozygous Kcnj13 null mice die within the first-day post-birth [41,42,43], mice with RPE-specific conditional Kcnj13 knockout and those with mosaic Kcnj13 expression in the RPE accurately reproduce the severe and progressive degeneration of photoreceptors seen in patients with KCNJ13 mutations [41, 44]. Notably, photoreceptors adjacent to RPE cells lacking Kir7.1 undergo degeneration, whereas those next to Kir7.1-expressing cells can survive, highlighting the critical role of this channel in maintaining photoreceptor integrity and survival [41]. Further mechanism studies on the Kcnj13 mutant zebrafish (obelixtd15, c.502T>C [p.F168L]) revealed impaired phagocytosis, abnormally distributed melanosome, and increased mitochondrial size and number in RPE cells [36].
hPSC-RPE cells have significantly accelerated research into the pathogenetic mechanisms for LCA16. The first LCA16 patient-specific hiPSC-RPE cells was obtained from a 10-year-old boy with a homozygous nonsense mutation (p.W53X) of KCNJ13 [5, 20]. The cells exhibited normal morphology with significantly decreased Kir7.1 activity and impaired POS phagocytosis [5]. Besides, Ying et al. generated Rab28−/− mouse and showed that Kir7.1 may facilitate POS phagocytosis in murine RPE cells by combining with Rab28 [45]. Yuki et al. demonstrated a direct link between Kir7.1 and phagocytic function in RPE cells. Their study revealed impaired phagocytosis in KCNJ13−/− hiPSC-RPE cells accompanied by downregulation of phagocytosis-related genes compared to wild-type controls [38]. In addition, they found that the KCNJ13−/− hiPSC-RPE cells exhibits enlarged cell size, disrupted cell alignment, broken cell surfaces, and heightened sensitivity to oxidative stress [38, 46]. These findings deepen our understanding of the mechanisms driving photoreceptor degeneration in patients with KCNJ13 mutations.
Notably, preclinical therapeutic research on LCA16 hiPSC-RPE cells has made significant progress. Since LCA16 is an autosomal recessive inherited disease, restoring partial expression of wild-type Kir7.1 through read-through compounds, such as neuroblastoma 84 (NB84), or via virus-mediated gene augmentation has significantly rescued impaired Kir7.1 currents and phagocytic function in hiPSC-RPE cells [5]. Recently, Kabra et al. successfully and efficiently corrected the p.W53X mutation using silica nanocapsules carrying adenine base editor 8e mRNA and sgRNA in LCA16 hiPSC-RPE cells and LCA16 mouse model (Kcnj13W53X/+ΔR). This treatment preserves normal vision function and retinal structure in treated mice [47]. Collectively, these preclinical studies have demonstrated the strong potential of gene therapy for treating LCA16. However, given the early onset of this disease, timely therapeutic intervention is crucial to achieving optimal clinical outcomes.
Chloride channels
Chloride, the most prevalent permeable anion in vivo, is mediated by various chloride channels, including chloride ligand-gated channels (CLC), chloride intracellular ion channels (CLIC), calcium-activated chloride channels (CaCC), and cystic fibrosis transmembrane conductance regulator (CFTR). Previous studies have demonstrated that Bestrophin-1 [6], anoctamin families [48, 49], CLC-2 [21], CLIC4 [50] and CFTR [51] are expressed in RPE cells. These chloride channels combined with sodium-potassium-chloride cotransporter (NKCC) maintain hydro-ionic homeostasis by facilitating chloride transepithelial transport from the subretinal space into the choroid [52]. Additionally, chloride channels are essential for critical RPE cell functions, such as POS phagocytosis and VEGF/PEDF secretion [6, 21, 52]. Consequently, understanding the expression and function of chloride channels within RPE cells is essential for unraveling their contributions to retinal health and disease. Herein, we review findings from previous research on chloride channels in hPSC-RPE cells, focusing on disease pathogenetic mechanisms and treatments related to channelopathies.
Bestrophin-1
Overview of BEST1 and bestrophinopathies
BEST1, also known as vitelliform macular dystrophy 2 (VMD2), encodes Bestrophin-1, an integral membrane protein predominantly expressed in the basolateral membranes of RPE cells [53]. Functionally, Bestrophin-1 forms a homopentameric protein complex and is the primary calcium-activated chloride channel (CaCC) in RPE cells [54, 55]. Bestrophin-1 regulates the POS phagocytosis and transepithelial Cl-Â transport [56]. Its mutations are related to various bestrophinopathies, including adult-onset vitelliform macular dystrophy (AVMD), best vitelliform macular dystrophy (BVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC) and retinitis pigmentosa 50 (RP50) [57, 58]. However, the diagnosis of RP50 remains controversial, as patients often present with additional pathogenic mutations in genes other than BEST1, which may also contribute to the RP phenotype [59, 60]. Additionally, ADVIRC can easily be misdiagnosed as RP since both conditions share a subset of similar clinical features, such as peripheral circumferential hyperpigmentation [60, 61]. Although several bestrophinopathies cases exhibit typical clinical characteristics, including the decreasing light peak of electrooculogram and specific egg yolk-like lesions, it remains difficult to differentiate all forms of bestrophinopathies from other types of IRD based solely on clinical features, thus necessitating genetic diagnosis.
Advances in disease model and pathogenetic mechanism of bestrophinopathies
Developing more suitable disease models is crucial for advancing our understanding of the diagnosis, treatment, and pathogenesis of bestrophinopathies. Canine multifocal retinopathy (cmr) mirrors bestrophinopathies both genetically and clinically. To date, three naturally occurring BEST1Â biallelic mutated canine models have been identified: cmr1 with a stop mutation (p.R25X), cmr2 with a missense mutation (p.G161D), and cmr3 with a frameshift mutation (p.P463fs) [62,63,64]. These models display recessive inheritance patterns, while most bestrophinopathies cases exhibit dominant inheritance, restricting their applicability. In addition to canine models, mice have also been employed to model bestrophinopathies. However, two independently created Best1 KO mice did not exhibit significant retinal abnormalities, except for the reduced light peak of electrooculogram [65, 66]. Although the Best1 p.W93C knock-in mouse displayed pronounced retinal detachments and subretinal debris deposition. The mouse maintained normal chloride currents in RPE cells, an unexpected finding considering the impairment of Bestrophin-1 [67]. Additionally, another knock-in mouse carrying the Best1 p.Y227N mutation displayed no significant ocular abnormalities even up to 2 years of age [68]. These findings suggest that mouse models face challenges in recapitulating the phenotypes of bestrophinopathies in humans.
The development and application of hPSC-RPE cells can significantly alleviate the limitations of traditional animal models, providing a more flexible platform for disease research. Various bestrophinopathies patient-specific hiPSC-RPE cells have been successfully generated. These cells showed the disruption of RPE specific functions, including defects in POS phagocytosis, accumulation of lipofuscin, and reduced transepithelial fluid transport [66, 69,70,71,72,73,74,75,76]. More specifically, Singh et al. found that the BEST1 mutations (p.A146K and p.N296H) impaired POS phagocytosis, possibly due to disturbances in protein degradation pathways, including decreased free-ubiquitin levels, increased protein oxidation, and disturbed exosome secretion rates in patient hiPSC-RPE cells [74, 75]. Lee et al. revealed that the ARB hiPSC-RPE cells carrying p.L40P and p.A915V mutations exhibited decreased transcellular fluid transport and significant differences in epithelial-mesenchymal transition gene profiles and tumor necrosis factor alpha (TNF-α) signaling compared to normal control or BVMD hiPSC-RPE cells with p.G96A mutation [76]. Furthermore, Volume-regulated anion channel (VRAC)-mediated currents were significantly reduced in hiPSC-RPE cells with p.A243V or p.Q238R mutations, indicating the crucial role of Bestrophin-1 in intracellular volume regulation [66].
Mutational effects on subcellular location and function of Bestrophin-1
Different mutations in Bestrophin-1 result in diverse functional outcomes, including loss of function (LOF) [72, 73, 77], gain of function (GOF) [78, 79], and dominant negative (DN) effects [79, 80]. Stephen et al. generated numerous hiPSC-RPE cells from BVMD patients with BEST1 mutations (p.A10T, p.R218H, p.L234P, p.A243T, p.Q293K, and p.D302A) and from ARB patients with p.I201T and p.P274R mutations. These mutations caused LOF by impairing calcium-dependent chloride currents [72, 73, 77]. Notably, p.366fsX18 mutation significantly enhanced chloride currents in hiPSC-RPE cells derived from ARB patients [78]. Similarly, hiPSC-RPE cells from an ADVIRC patient with p.V86M mutation exhibited increased chloride currents [71]. These GOF effects have also been observed in HEK293 cells overexpressing various mutant Bestrophin-1, including p.D203A, p.I205T, and p.Y236C [81].
Some mutations also affect the subcellular localization of Bestrophin-1 [82]. Herein, we focus on the change in hPSC-RPE cells. The p.V235A mutant Bestrophin-1 is abnormally expressed at the apical and basolateral membranes of hiPSC-RPE cells from ADVIRC patients [69]. Cordes et al. demonstrated that transfecting normal hiPSC-RPEs with the p.T6P mutant BEST1 could significantly decrease the membrane localization of both Bestrophin-1 and CaV1.3 [83]. Moreover, mutations, such as p.R141H or p.A195V, significantly decreased Bestrophin-1 expression compared with the controls [6]. Interestingly, some heterozygous mutations (p.A10T, p.R218H, p.L234P, p.A243T, p.Q293K, p.D302A) caused allelic expression imbalance (AEI) in patient-specific hiPSC-RPE cells, displaying a significantly higher mRNA expression of the mutated allele than the wild-type BEST1 allele, indicating a DN effect at the transcriptional level [79, 80]. The abnormalities in the expression, subcellular localization, and channel activity of Bestrophin-1 reflect the varied inheritance patterns and the diverse clinical phenotypes in bestrophinopathies. These complexities also indicate the challenges in developing an univeral, effective treatment for all disease subtypes.
Innovative therapy strategies for bestrophinopathies
Developing effective treatments for bestrophinopathies remains a compelling target, because they are one of the most prevalent form of retinal channelopathies. Moreover, bestrophinopathies progress slowly, enabling many patients to retain basic visual functions into their later decades, thus providing a substantial window for therapeutic intervention [80, 84]. Additionally, this disorder is mainly caused by the disruption of RPE cells, which are strategically positioned as a single layer between the neural retina and the choroidal capillaries. This anatomical arrangement facilitates drug delivery via intravitreal, subretinal, or choroidal routes.
Small molecule drugs are considered promising and cost-effective options for treating bestrophinopathies. Specifically, bafilomycin-A1 slows down POS degradation in BVMD hiPSC-RPE cells, whereas valproic acid accelerates POS degradation [75]. Lin et al. found that curcumin could significantly enhance the expression of ZO-1 and Bestrophin-1 compared with other candidate drugs (vitamin C, β-carotene, retinoic acid, forskolin, isoproterenol, Q10), thus improving intracellular reactive oxygen species and promoting phagocytosis functions in BVMD hiPSC-RPE cells [85]. Liu et al. demonstrated that 4PBA (sodium phenylbutyrate), an FDA-approved drug for urea cycle disorders, and its analogue 2-NOAA (2-naphthoxyacetic acid), could rescue the expression, localization, and channel activity of Bestrophin-1 in BVMD or ARB patient-specific hiPSC-RPE cells [86]. However, the effective therapeutic concentrations of 4PBA and 2-NOAA are extremely high (2.5 mM) for practical treatment [86, 87]. Recent studies have discovered that 25 µM of tadalafil, an alternative to 4PBA, can effectively restore chloride current in HEK293T cells overexpressing p.M325T mutation of Bestrophin-1 [87]. However, tadalafil is ineffective for the p.R141H and p.L234V mutations, highlighting the complexity of bestrophinopathies [87].
Compared with small molecule drugs, gene therapy appears to have greater potential for treating bestrophinopathies. Gene augmentation therapy works by expressing an exogenous normal protein corresponding to the mutated protein within cells to rescue cellular function. BEST1 encodes only 585 amino acids [53], indicating that it can easily be encapsulated into adeno-associated virus (AAV), making it highly suitable for this treatment approach. Studies have shown that various BEST1 mutations, which lead to LOF or DN effects, can be effectively addressed by expressing exogenous normal Bestrophin-1. This method restores chloride current and POS phagocytosis in hiPSC-RPE cells, regardless of whether the inheritance pattern is AD or AR [6, 73, 82, 88]. However, some autosomal dominant genetic bestrophinopathies are resistant to gene augmentation, possibly due to the heightened toxicity associated with BEST1 mutations, where the expression of exogenous normal copies cannot sufficiently counteract the negative effects [6, 79]. Gene editing is used in such cases to silence the mutated BEST1 allele. For instance, CRISPR/Cas9 gene editing, specifically targeting the p.A146K mutation in BEST1, can restore chloride current and improve POS digestion function in patient-specific hiPSC-RPE cells [6]. Gene editing can also be used for GOF mutations in bestrophinopathies. Moreover, non-selective CRISPR/Cas9-mediated gene silencing combined with gene augmentation can be used to treat bestrophinopathies. Recent studies have demonstrated that silencing both alleles of BEST1 in hiPSC-RPE cells and expressing exogenous normal BEST1 can effectively rescue the Bestrophin-1-mediated chloride current [79]. Notably, this approach non-discriminately silences both alleles of BEST1, theoretically making it applicable to nearly all bestrophinopathies patients. Nonetheless, the efficiency and potential off-target effects of gene editing should be further evaluated.
CLC-2
CLC-2 and retinal degeneration
Chloride voltage-gated channel 2 (CLC-2), a member of the CLC family, is a two-pore homodimeric voltage-gated chloride channel. It was first cloned from the rat heart and brain in 1992 and was subsequently found to be widely expressed in mammalian cells, especially the epithelial cells [89, 90]. CLC-2 participates in the regulation of cell volume, stabilizing membrane potential, facilitating transepithelial chloride transport, and aiding in signal transduction [90, 91].
Mutations in CLCN2 can lead to various disorders, including leukoencephalopathy with ataxia (LKPAT; MIM#615651) [92, 93], epilepsy (though this remains controversial) [94, 95], familial hyperaldosteronism type 2 (HALD2; MIM#605635) [96, 97], infertility [98] and retinal degeneration [21, 92]. In previous studies, ocular defects were reported in 6 of 15 LKPAT patients carrying homozygous CLCN2 mutations. More specifically, Depienne et al. initially observed low vision and visual field defects in LKPAT patients carrying CLCN2 mutation (p.W570X or p.L144_I145del) without detailed phenotypic descriptions or objective examinations [92]. Bella et al. also detected abnormal ERG and VEP responses in a patient with p.G503R mutation. However, the patient maintained a normal fundus and visual field, suggesting the retinal disorder is mild [98]. Another study detected presbyopia and normal far-sight in a patient with p.H590P mutation. However, the nearly normal fundus appearance did not corroborate the CLCN2 mutation as the cause of these ocular abnormalities [99]. Our studies identified an LKPAT patient with homozygous p.R753X nonsense mutation [21, 100]. This patient showed severe bilateral retinal degeneration involving loss of photoreceptors and RPE cells in the central macula, diffuse chorioretinal atrophy, peripheral retinal bone spicule pigmentation, and bilateral microvascular leakage [21]. Most recently, Cheng et al. reported another LKPAT patient with a novel CLCN2 mutation (p.P461Lfs*13). The patient displayed bilateral blurred vision, posterior capsular cataracts, macular retinal atrophy, peripheral retinal pigmentation, and a novel sign of vacuole-like vitreoretinopathy [101]. Collectively, these cases suggest that retinal degeneration associated with CLCN2 mutations should be classified as a novel autosomal recessive IRD (CLCN2-IRD), which may aid in the identification of similar cases.
Mutant CLC-2 disrupts critical functions of RPE cells
Genetically engineered mouse models can recapitulate the clinical features of CLCN2 mutation-mediated retinal disorders. Mice with LOF mutations in CLC-2, including Clcn2nmf240 [102], Clcn2nmf289 [103], Clcn2tm1Mlv [104], and Clcn2tm1Tjj [105], exhibit progressive retinal degeneration, leukodystrophy and azoospermia. The retinal disorders persist even when Clcn2 is conditionally knocked out in murine RPE cells [106]. Furthermore, targeted expression of CLC-2 in RPE cells can rescue photoreceptor degeneration in Clcn2 KO mice, indicating the critical role of RPE cells as the primary site of the retinal degeneration [107]. Nevertheless, the localization of CLC-2 in RPE cells remains contentious. Ectopic HA-tagged CLC-2 is specifically localized in the apical membrane of RPE cells of Clcn2 KO mice, aligning with subcellular locations observed in human fetal RPE (hfRPE) and ARPE-19 cells [107]. Recent studies have found the same subcellular location of CLC-2 in hiPSC-RPE cells [21]. However, Mamaeva et al. showed distinct subcellular locations of CLC-2 in hiPSC-RPE cells (basolateral membrane and apical microvilli). They also showed that CLC-2 was highly expressed at the apical pole of fluid-filled domes [29]. CLC-2 has also been reported in both cell center and intercellular junction of hESC-RPE cells [108]. The variability in the expression and localization of CLC-2 may be influenced by the state of RPE cells and culture conditions used (conventional multi-well plates vs. Transwell inserts). In Transwell inserts, fluid does not accumulate beneath RPE cells, while a subset of RPE cells with active apical-basal fluid transport in culture plates forms fluid-filled domes.
CLCN2 mutations affect essential RPE cell functions, particularly POS phagocytosis. Mamaeva et al. showed that CLC-2 inhibitor (GaTx2) could impede POS phagocytosis in hiPSC-RPE cells [29]. Similarly, CLCN2 p.R753X mutation impairs POS phagocytosis in patient-specific hiPSC-RPE cells [21]. POS is ingested by the RPE cells forming a motile organelle, phagosome, which is digested by interacting with endosomes and lysosomes [109]. In this process, CD36 plays a crucial role in mediating POS ingestion [110]. Microtubule-based motor transport is essential for the timely degradation and clearance of POS [109]. However, both processes can be disrupted in patient hiPSC-RPE cells, indicating that defects in phagocytosis promote the initiation and progression of CLCN2-IRD [21]. Besides POS phagocytosis of RPE in retina, the neuroglia (oligodendrocytes and astrocytes) in the brain and Sertoli cells in the testis play critical roles in the phagocytosis of myelin debris [111, 112] and apoptotic germ cells [113], respectively. Targeted deletion of CLCN2 in these cells promote leukodystrophy and testicular degeneration [106], suggesting that impaired phagocytosis may be a common underlying mechanism in the varied clinical manifestations observed in patients with CLCN2 mutations.
CLC-2 also plays an important role in maintaining the integrity of the tight junction (TJ) barrier, the length and arrangement of apical microvilli, and the secretion of VEGF/PEDF in RRE cells. Studies have shown that transdermal current and voltage are decreased in the RPE cells of Clcn2 KO mice as the tight junction (TJ) protein ZO-1 is accumulated at branch points [105, 107]. CLC-2 overexpression enhances the TJ barrier and increases the expression of occludin in human intestinal Caco-2 cells [114]. However, the hiPSC-RPE cells in patient with CLCN2 p.R753X mutation maintain the normal transepithelial resistance, permeability of FITC-dextran, and expression of occludin, as observed in the epithelia of the small intestine and colon of Clcn2 KO mice [21, 115]. Besides the disruption of TJ barrier, highly disarranged and elongated microvilli have been observed in RPE cells of Clcn2 KO mice. However, it can be rescued by exclusive expression of normal CLC-2 in these cells [102, 107]. Nonetheless, the patient hiPSC-RPE cells have similar distributions, morphologies, and lengths for their apical microvilli compared with normal control [21]. These may be due to the fact that the p.R753X mutant CLC-2 is still inserted into the cell membrane and retains a weak chloride current [21]. As for the secretion of VEGF and PEDF, it is significantly decreased by the CLC-2 inhibitor GaTx2 in hiPSC-RPE cells, suggesting a potential impact on the choroid and neural retina [29].
Collectively, these findings partially support the hypothesis that the disruption of RPE cells may the primary cause of retinal degeneration in CLCN2-IRD. Additionally, it is noteworthy that retinal disorder has not been observed in HALD2 patients or mice carrying CLC-2 GOF mutations [90, 91, 109].
Feasible treatment approaches for CLCN2-IRD
Previous study shows that the photoreceptor degeneration in Clcn2 KO mice can be effectively rescued by targeted expression of normal CLC-2 in RPE cells [106]. In our study, two isogenic hiPSC-RPE cell lines were developed using CRISPR/Cas9 technology to correct one allele of the CLCN2 p.R753X mutation. Both corrected hiPSC-RPE cells showed normal chloride currents and POS phagocytosis [21], illustrating that partial restoration of CLC-2 function can maintain the overall functionality of these cells. These findings suggest that gene therapy is a promising treatment strategy for CLCN2-IRD.
Other chloride channels
Despite the presence of other chloride channels, such as CFTR, CLIC4, Anoctamin-1 and Anoctamin-2 in RPE cells [48,49,50, 116, 117], research on these channels remains sparse. Moreover, Xu et al. analyzed anion conductance in murine RPE cells, hinting at the existence of additional, yet-to-be-identified chloride channels [118]. The hPSC-RPE cells offer a promising platform to investigate the expression, subcellular localization, and function of these chloride channels in RPE cells.
CFTR is a 1480 amino acid-long membrane protein and anion channel belonging to the ABC transporter superfamily. It is expressed in various tissues, including the lung, liver, intestine, sweat gland, reproductive tract, and retina [116, 119]. Therefore, mutations in CFTR can lead to multisystem diseases, such as cystic fibrosis (MIM#219700), congenital bilateral absence of vas deferens (MIM#277180) and hereditary pancreatitis (MIM#167800). Apart from modulating chloride conductance, CFTR regulates the activity of other channels, fluid transport, and lipid metabolism [119]. In hfRPE cells, CFTR is found to be expressed on the apical and basolateral membranes [52, 116]. In hESC-RPE cells, CFTR is concentrated in ring-like structures, but its subcellular localization is not well known [120]. Previous research has demonstrated that CFTR regulates fluid transport and reacidification of alkalinized lysosomes [51, 121]. Furthermore, Cftr KO mice show decreased response in RPE-driven ERG components [122]. However, retinal disorders have not been reported in patients carrying CFTR mutations. Over the last decade, few studies have explored the CFTR in RPE cells and future investigations are warranted to uncover its roles and mechanisms.
CLIC4 is a multifunctional protein belonging to the CLIC family. It exerts chloride channel activity in synthetic lipid bilayers and modulates several intracellular processes such as cell proliferation, apoptosis, apical membrane biogenesis, cytoskeletal remodeling, and endosomal sorting [123, 124]. In rat RPE cells, CLIC4 is highly expressed in the apical microvilli. Silencing it impairs apical microvilli, basolateral infoldings of RPE cells, and reduces retina-RPE adhesion [125]. In human RPE cells, CLIC4 co-localizes with matrix metalloproteinase 14 (MMP14) in the late endosomes of ARPE19 cells. Downregulation of CLIC4 decreases the secretion of MMP14 and MMP2, thereby blocking the degradation of the extracellular matrix (ECM) [50]. Although CLIC4 is also expressed in hESC-RPE cells, its subcellular localization has not been reported [50]. Knockdown of CLIC4 disrupts the cell morphology and decreases the apical secretion of MMP2 in polarized hESC-RPE cells [50]. It has been reported that MMP2 and MMP14 regulate the ECM and might be related to AMD [126, 127]. A novel mouse model with RPE-specific Clic4 KO exhibited major functional and pathological hallmarks of dry AMD [117]. This conditional KO reduced the expression of MMP2 and MMP3 in RPE cells, while no changes were observed in MMP14 [117]. Alterations in MMPs expression may contribute to Bruch’s membrane remodeling, potentially promoting the development of drusen, a hallmark of dry AMD. Given the multifaceted role of CLIC4, current research on its effects in RPE cells remains insufficient. Future studies using both animal models and hPSC-RPE cells are essential for comprehensively elucidating the role of CLIC4 in RPE cells and its involvement in AMD pathogenesis.
Sodium channels
Sodium channels include two major classes: the epithelial sodium channel (ENaC) and the voltage-gated sodium channel (VGSC or NaV). ENaCs are heterotrimeric proteins predominantly found in polarized epithelial cells. They are key regulators of cellular ion and water homeostasis. Certain ENaC subunits have been detected in human donor RPE cells, indicating their potential roles in sodium transepithelial transport [52, 128]. Currently, the expression and function of ENaC in hPSC-RPE cells has not yet been investigated. In contrast, recent studies have revealed the expression and function of NaV in hESC-RPE cells [4]. This section primarily focuses on the exploration of NaV channels in hPSC-RPE cells.
Voltage-gated sodium channel (NaV)
NaV consists of an alpha subunit and one or two beta subunits, with the alpha subunit alone being sufficient for channel activity [129]. Further, the alpha subunit contains ten subtypes, ranging from NaV1.1 to NaV1.9, and NaX [130]. They are considered as the hall marks of various excitable cells like neurons, glial cells, and muscles, contributing to the formation of an action potential [131]. RPE cells are classified as non-excitable ones, leading to ongoing debate regarding the expression and function of NaV in these cells. Electrophysiological studies show that sodium currents are undetectable in mature RPE cells and are only observed in cultured RPE cells. This discrepancy suggests that the presence of these currents may be result from neuroepithelial differentiation associated with in vitro culture conditions [52, 132]. Lidgerwood et al. found no detectable sodium currents in hESC- or hiPSC-derived RPE cells using an innovative one-step differentiation protocol [133]. However, recent research has challenged this view, with findings that the NaV expression, subcellular localization, and function are detected in both hESC-RPE cells and freshly isolated murine RPE cells [4]. Immunostaining revealed the presence of NaV1.1 and NaV1.3 to NaV1.9 on the apical membrane and/or cell-cell junctions of hESC-RPE cells (Fig. 2 and Suppl. Table 1). During the first nine days of maturation, NaV became more specifically localized to cell-cell junctions (NaV1.4) and the apical membrane (NaV1.5 and NaV1.8), indicating their potential as indicators of RPE cell maturation and polarization [4].
Sodium and calcium channels in hPSC-RPE cells. The subcellular localization of sodium channels (NaV1.1, NaV1.3 to NaV1.9) and calcium channels (CaV1.1 to CaV1.3, CaV3.1 to CaV3.3) in hPSC-RPE cells. Despite functional studies of TRPML1 in hPSC-RPE cells reported, its localizations in hPSC-RPE cells have not been studied; thus, the subcellular localization is deduced from its locations in ARPE19 cells. Abbreviations: sER, smooth endoplasmic reticulum; rER, rough endoplasmic reticulum; GC, Golgi complex; Mit, mitochondrion
The currents of various NaV have been recorded in hESC-RPE cells by whole-cell patch-clamp. These currents were confirmed through the application of sodium channel blockers (TTX and QX-314), as well as selective blockers of NaV subtypes (4,9-anhydro-TTX for NaV1.6, A-803467 for NaV1.8 and μ-Conotoxin GIIB for NaV1.4) [4]. Furthermore, inhibiting these NaV by blockers or shRNA suppresses the POS phagocytosis, particularly during internalization and processing [4]. Recent study has demonstrated that sodium based voltage spikes in hESC-RPE cells can spread laterally in the monolayer through gap junctions, revealing a novel potential role of RPE cells [134]. Although these findings are compelling, the absence of independent validation from other researchers highlights the need for further studies to draw definitive conclusions.
Calcium channels
Calcium ions, as critical second messengers, regulate various cellular processes in RPE cells [52]. Multiple calcium channels modulate intracellular calcium level. Mutations in these channels have been linked to several retinal diseases, including AMD and bestrophinopathies [56, 83, 135]. However, the current understanding of calcium channels in RPE cells remains incomplete. Inconsistencies among existing findings likely stem from the intricate and sensitive nature of calcium signaling in these cells [3, 29]. In this context, we aim to provide a comprehensive review of current research on calcium channels in hPSC-RPE cells, highlighting the advances made to date and the critical gaps that warrant further investigation.
Voltage-dependent calcium channels (VDCCs), also known as voltage-gated calcium channels (VGCCs), selectively allow the passage of calcium ions when activated by depolarizing membrane potentials. These channels comprise at least 10 different subtypes, including L-type channels (CaV1.1-1.4), P/Q-type channel (CaV2.1), N-type channel (CaV2.2), R-type channel (CaV2.3), and T-type channels (CaV3.1-3.3) [52].
CaV1.3
Subcellular location of CaV1.3
Three L-type calcium channels (CaV1.1, CaV1.2, and CaV1.3) are expressed in RPE cells, among which CaV1.3 is considered to play a primary role [52, 56, 132, 136, 137]. The activity of CaV1.3 is regulated through phosphorylation, which shifts its voltage-dependent activation toward more negative voltage, enhancing its activity at the resting membrane potential of RPE cells [56]. In vivo, CaV1.3 is localized at the basolateral membrane of murine RPE cells [138]. In contrast, in vitro studies show that it is present on both the apical and basolateral membranes of cultured murine RPE cells [3]. In hESC-RPE cells, the expression pattern of CaV1.3 changes as the cells mature. Initially, on the first day post-confluence, CaV1.3 is distributed broadly across the cell. As the cells adopt a more epithelioid and eventually cobblestone morphology, CaV1.3 becomes increasingly concentrated on both the apical and basolateral sides. This localization includes distinct apical puncta that co-localize with pericentrin. However, as the cells continue to mature and develop pigmentation, detection of CaV1.3 at the basolateral membrane becomes challenging, while the apical localization becomes more homogeneous and pronounced [3]. This dynamic subcellular localization indicates that CaV1.3 may serve as a biomarker for the maturity and quality of hPSC-RPE cells.
Role of CaV1.3 in retinal diseases
CaV1.3 has been shown to participate in the pathological mechanism of various retinal diseases, such as AMD and bestrophinopathies [56, 83, 135]. Research on hPSC-RPE cells has significantly advanced our understanding of the role of CaV1.3 in these disorders. In AMD patient-specific hiPSC-RPE cells, CaV1.3 exhibits a greater shift toward intracellular compartments compared to control cells, which may alter calcium signaling and indirectly disrupt key molecular pathways related to inflammasome activation and autophagy in AMD pathology [135]. Choroidal neovascularization (CNV) represents the most visually threatening clinical phenotype for AMD. Abnormal secretion of VEGF and PEDF is vital for the development of CNV. VEGF promotes the formation of new blood vessels, while PEDF inhibits neovascularization. Research on hiPSC-RPE cells has shown that blocking L-type calcium channels with nifedipine significantly decreases basolateral secretion of VEGF [29]. Further research has confirmed the effects of nifedipine and demonstrated that treating hESC-RPE cells with the L-type channels activator (-) BayK8644 increases the secretion of VEGF [3]. In addition, nifedipine also significantly decreased PEDF secretion on the basolateral side of hiPSC-RPE cells [29]. In AMD patient-specific hiPSC-RPE cells, apical PEDF was significantly reduced than the control, although the basolateral secretion was unchanged [135]. These observations highlighting the pivotal role of CaV1.3 in the regulation of VEGF and PEDF secretion and development of wet AMD [139].
CaV1.3 also contributes to the development of bestrophinopathies. The studies indicate that POS phagocytosis is commonly disrupted in bestrophinopathies. In CaV1.3 KO mice, it was found that CaV1.3 mediated the circadian regulation of POS phagocytosis, exhibiting electrophysiological characteristics similar to those observed in bestrophinopathies patients [56, 140]. Manipulating L-type channels activity in hESC-RPE cells impaired POS phagocytosis, regardless of inhibiting or activating these channels [3]. Furthermore, Cordes et al. demonstrated that mutant Bestrophin-1 (p.D302A) decreased its interaction with CaV1.3, thereby decreasing the calcium channel activity and altering membrane localization [83]. This disruption affects calcium signaling in hiPSC-RPE cells, highlighting the essential role of CaV1.3 in the development of bestrophinopathies [83].
Other L-type calcium channels
In addition to CaV1.3, CaV1.1 and CaV1.2 have been described in hPSC-RPE cells. CaV1.2 is expressed on the basolateral membrane of hESC-RPE cells, while CaV1.1 has not been detected [3]. Mamaeva et al. found that CaV1.1 and CaV1.3 were located in the basolateral membrane in hiPSC-RPE cells [29]. In contrast, Karema-Jokinen et al. reported that all three L-type channels were detectable in hiPSC-RPE cells [135]. The differences in expression and subcellular localization of these channels might be influenced by cell type and culture conditions. Recent studies have uncovered a novel function of L-type calcium channels in RPE cells: downregulation of these channels with nifedipine enhances gap junctional conductance in hESC-RPE cells [134].
T-type calcium channels
Although seldom discussed, T-type channels are also expressed in RPE cells [3, 136]. In primary human RPE cells, the mRNA level of CaV3.1 and CaV3.3 was detected by RT-PCR [136]. In hESC-RPE cells, fast transient T-type resembling currents are detectable [3]. The CaV3.1 and CaV3.2 are detected in murine RPE cells and hESC-RPE cells, whereas CaV3.3 is not detectable by immunofluorescent staining [3]. It has been shown that CaV3.1 is specifically localized on the apical membrane of hESC-RPE cells and in the entire cell membrane of murine RPE cells. CaV3.2 is expressed on both the basolateral membrane and cell-cell junctions of these cells [3]. In hiPSC-RPE cells, CaV3.1 exhibits a similar distribution, while CaV3.3 is also detectable on the apical membrane [29]. These channels regulate the POS phagocytosis, and inhibition of the T-type channels with ML218 in hESC-RPE cells enhances the binding and internalization of POS [3], whereas another T-type channel inhibitor, TTA-A2, reduces the internalized number of POS in hiPSC-RPE cells [29].
Overall, VDCCs are essential for hPSC-RPE cells to perform their functions. However, the regulatory mechanisms of these ion channels in RPE cells are complex and multifaceted. For example, both activators and inhibitors of L-type channels have been shown to impair POS phagocytosis [3]. This intricate regulation may involve negative feedback mechanisms and the interplay of other ion channels, including maxi-K, Kir7.1, Bestrophin-1, and CLC-2 [5, 21, 29, 83, 140]. Furthermore, since hPSC-RPE cells express multiple L-type and T-type calcium channels, the functional loss of one calcium channel could potentially be compensated by others within the same family, as observed in mouse basal forebrain neurons [141]. Altogether, the complex regulatory mechanisms of VDCCs in hPSC-RPE cells necessitate further investigation.
Transient receptor potential (TRP) channels
Transient receptor potential (TRP) channels, comprising 28 members, are classified into seven subfamilies including TRPA, TRPC, TRPM, TRPML, TRPN, TRPP, and TRPV. These channels regulate calcium signaling and various functions of RPE cells. A recent comprehensive study by David et al. summarized the recent advances for these channels in retina [142]. In this section, we focus on the roles of TRPML1 in RPE cells.
Transient receptor potential mucolipin 1 (TRPML1), encoded by the MCOLN1, is expressed on the endo-lysosomal membrane across almost all mammal cells, where it transduces calcium signals and lysosomal homeostasis [143, 144]. It is involved in diverse cellular processes such as lysosomal exocytosis, lipid trafficking, and autophagy [145]. Mutations in TRPML1 cause mucolipidosis type IV (MLIV), a severe neurological lysosomal storage disorder characterized by psychomotor retardation and ophthalmologic abnormalities [146]. In Mcoln1 KO mouse, photoreceptor degeneration and optic nerve pathology have been detected [147]. Recent studies found that treatment of hESC-RPE cells, ARPE19 cells, and murine RPE cells with the TRPML agonist ML-SA1 significantly increased cytoplasmic calcium levels. The increase originates from lysosomal stores rather than the endoplasmic reticulum or extracellular space. Moreover, this TRPML1-mediated response can be diminished by accumulation of lysosomal lipid [148, 149]. These findings shed light on the pathogenetic mechanism underlying MLIV and highlight the critical role of TRPML1 in RPE cells. Besides TRPML1, other TRP channels such as TRPV2, TRPV4, TRPV5, TRPV6, TRPM1, TRPM3, TRPM7, TRPC1, and TRPC4 have not comprehensively been investigated in hPSC-RPE cells [142]. Understanding their functions may provide new insights into retinal disease mechanisms and potential therapeutic targets.
General summary
In this review, we summarize the current research on ion channels in hPSC-RPE cells (Figs. 1 and 2 and Suppl. Table 1). These studies have significantly advanced the clinical applications of hPSC-RPE cells. On one hand, they have directly facilitated research on the pathogenic mechanisms and therapies for channelopathies. On the other hand, they have contributed to the development of more stable and mature RPE cells for preclinical and clinical studies in RPE cell transplantation.
Dysfunction of bestrophin-1, CLC-2, and Kir7.1 channels in RPE cells can lead to bestrophinopathies, CLCN2-IRD, and LCA16, respectively [20, 21, 40, 57, 58, 92]. Patient-specific hiPSC-RPE cells with these channelopathies exhibit impaired RPE functions, characterized by reduced ion channel conductivity and diminished POS phagocytosis. Gene therapies, including gene augmentation and gene editing, have demonstrated promising therapeutic effects in these hiPSC-RPE cells [5, 6, 21]. Moreover, the blood-retinal barrier reduces the immune response to AAV delivery. RPE cells, as a monolayer, are particularly amenable to AAV infection via intravitreal or subretinal injection [150]. Based on these findings, we believe that gene therapies for these channelopathy patients are safe and hold significant promise. However, clinical trials targeting RPE channelopathies are currently lacking, possibly due to the mild and slow progression of vision loss in many patients, which may diminish the need for such trials. Additionally, the rarity of channelopathies such as CLCN2-IRD may limit the commercial viability of developing gene therapy [21]. To facilitate the clinical translation of treatments for these channelopathies, it is essential to optimize current gene therapy strategies to enhance safety, efficacy, and cost-effectiveness.
Since the initial use of hESC-RPE cells in treating AMD and STGD, both hESC- and hiPSC-derived RPE cells have been widely employed in clinical trials for retinal degenerative diseases [22, 23, 151, 152]. While delivery methods vary, using either RPE cell suspensions [22, 151] or cell patches [23, 153], studies consistently demonstrate the safety of hPSC-RPE cell transplantation. Notably, some patients have reported slight improvements in vision following hPSC-RPE transplantation, although surgical procedures and immunomodulation may also contribute to these positive outcomes [12, 153,154,155]. The physiological condition of hPSC-RPE cells is a major factor influencing the success of cell transplantation therapies. Treatments for AMD and STGD specifically target macular degeneration. Research indicates that RPE cells in the macula exhibit distinct cellular morphology and gene expression profiles compared to those in surrounding regions [156, 157]. Furthermore, the macula has a heightened demand for RPE functions, such as POS phagocytosis, N-retinylidene-N-retinylethanolamine (A2E) degradation, vitamin A recycling, and fluid regulation—all of which are closely linked to ion channel activity [3,4,5,6, 158]. By assessing ion channel expression and localization in hPSC-RPE cells, researchers may identify key biomarkers that indicate cell maturity and quality, thus improving the selection of cells for more effective cell transplantation therapy. For example, CaV1.3 may serve as a useful biomarker, as its subcellular localization is closely associated with the maturation of hPSC-RPE cells [3].
The hPSC-RPE cells provide a valuable platform for investigating ion channel activity, subcellular localization, expression, and function. However, several challenges persist. Firstly, the differentiation methods used to obtain hPSC-RPE cells from hESC or hiPSC vary widely, leading to inconsistencies in cell purity, maturity, and characteristics [159,160,161]. Furthermore, culture conditions—including media composition (e.g., FBS, B27, NEAA), materials (e.g., plates, chamber inserts, coverslips, amniotic membranes, Parylene C, Poly L-lactic-co-glycolic Acid), and coatings (e.g., Matrigel, laminin, poly-D-lysine) —significantly impact cell growth and polarization [162]. Additionally, as hPSC-RPE cells mature, they progressively acquire pigmentation, establish tight junctions, and attain robust polarity—features essential for POS phagocytosis, transepithelial transport and VEGF/PEDF secretion. However, these characteristics rapidly diminish with passaging, indicating that variations in the maturation stages of hPSC-RPE cells can result in differing experimental outcomes. This may explain discrepancies in the subcellular localization of VDCCs [3, 29, 135] and the expression of NaV in RPE cells [4, 133]. To ensure reliable results, it is crucial to maintain stringent hPSC-RPE cell culture conditions in future research.
Research on hPSC-RPE cells has greatly enhanced our understanding of ion channels in RPE cells, particularly Bestrophin-1, CLC-2, Kir7.1, and CaV1.3. Recent discoveries of NaV, KV, and maxi-K channels, along with evidence of electrical excitability and lateral signal propagation in hPSC-RPE cells, suggest novel functions beyond the traditional roles attributed to these cells [27, 29, 134]. Additionally, POS phagocytosis in hPSC-RPE cells is highly sensitive to the activity of various ion channels, including potassium, sodium, chloride, and calcium channels [3,4,5,6]. These findings underscore the urgent need for further in-depth research to elucidate the complex roles of ion channels in hPSC-RPE cells.
Data availability
The data supporting the conclusions of this article are all online.
Abbreviations
- AAV:
-
Adeno-associated virus
- ADVIRC:
-
Autosomal dominant vitreoretinochoroidopathy
- AEI:
-
Allelic expression imbalance
- AMD:
-
Age-related macular degeneration
- ARB:
-
Autosomal recessive bestrophinopathy
- AVMD:
-
Adult-onset vitelliform macular dystrophy
- A2E:
-
N-retinylidene-N-retinylethanolamine
- BVMD:
-
Best vitelliform macular dystrophy
- CaCC:
-
Calcium-activated chloride channel
- CFTR:
-
Cystic fibrosis transmembrane conductance regulator
- CLC:
-
Chloride ligand-gated channel
- CLC-2:
-
Chloride voltage-gated channel 2
- CLIC:
-
Chloride intracellular ion channel
- cmr:
-
Canine multifocal retinopathy
- CNV:
-
Choroidal neovascularization
- DN:
-
Dominant negative
- ECM:
-
Extracellular matrix
- ENaC:
-
Epithelial sodium channel
- GOF:
-
Gain of function
- HALD2:
-
Familial hyperaldosteronism type 2
- hESC:
-
Human embryonic stem cells
- hfRPE:
-
Human fetal RPE
- hiPSC:
-
Human induced pluripotent stem cells
- hiPSC-RPE cells:
-
hiPSC-derived RPE cells
- hPSC:
-
Human pluripotent stem cells
- hPSC-RPE cells:
-
hPSC-derived RPE cells
- K+ :
-
Potassium
- Kir channels:
-
Inwardly rectifying potassium channels
- KV :
-
Voltage-gated potassium channels
- LCA:
-
Leber congenital amaurosis
- LKPAT:
-
Leukoencephalopathy with ataxia
- LOF:
-
Loss of function
- maxi-K:
-
Large-conductance calcium-activated potassium channel
- MLIV:
-
Mucolipidosis type IV
- MMP14:
-
Matrix metalloproteinase 14
- MMP2:
-
Matrix metallopeptidase 2
- NaV :
-
Voltage-gated sodium channel
- NB84:
-
Neuroblastoma 84
- NF-κB:
-
Nuclear factor-kappa B
- NKCC:
-
Sodium-potassium-chloride cotransporter
- PEDF:
-
Pigment epithelium-derived factor
- POS:
-
Photoreceptor outer segments
- RP:
-
Retinitis pigmentosa
- RPE:
-
Retinal pigment epithelial
- STGD:
-
Stargardt disease
- SVD:
-
Snowflake vitreoretinal degeneration
- TJ:
-
Tight junction
- TNF-α:
-
Tumor necrosis factor alpha
- TRP channels:
-
Transient receptor potential channels
- TRPML1:
-
Transient receptor potential mucolipin 1
- VDCCs:
-
Voltage-dependent calcium channels
- VEGF:
-
Vascular endothelial growth factor
- VGCCs:
-
Voltage-gated calcium channels
- VGSC:
-
Voltage-gated sodium channel
- VMD2:
-
Vitelliform macular dystrophy 2
- VRAC:
-
Volume-regulated anion channel
- 2-NOAA:
-
2-naphthoxyacetic acid
- 4PBA:
-
Sodium phenylbutyrate
References
Kokkinaki M, Sahibzada N, Golestaneh N. Human induced pluripotent stem-derived retinal pigment epithelium (RPE) cells exhibit ion transport, membrane potential, polarized vascular endothelial growth factor secretion, and gene expression pattern similar to native RPE. Stem Cells. 2011;29(5):825–35.
Miyagishima KJ, et al. In pursuit of authenticity: Induced Pluripotent Stem cell-derived retinal pigment epithelium for clinical applications. Stem Cells Transl Med. 2016;5(11):1562–74.
Korkka I, et al. Functional voltage-gated calcium channels are Present in Human Embryonic Stem cell-derived retinal pigment epithelium. Stem Cells Transl Med. 2019;8(2):179–93.
Johansson JK, et al. Sodium channels enable fast electrical signaling and regulate phagocytosis in the retinal pigment epithelium. BMC Biol. 2019;17(1):63.
Shahi PK, et al. Gene Augmentation and Readthrough Rescue Channelopathy in an iPSC-RPE model of congenital blindness. Am J Hum Genet. 2019;104(2):310–8.
Sinha D, et al. Human iPSC modeling reveals mutation-specific responses to Gene Therapy in a Genotypically Diverse Dominant Maculopathy. Am J Hum Genet. 2020;107(2):278–92.
Wang Y, et al. Genetic and clinical features of BEST1-associated retinopathy based on 59 Chinese families and database comparisons. Exp Eye Res. 2022;223:109217.
Davidson AE, et al. Missense mutations in a retinal pigment epithelium protein, bestrophin-1, cause retinitis pigmentosa. Am J Hum Genet. 2009;85(5):581–92.
Sergouniotis PI, et al. Recessive mutations in KCNJ13, encoding an inwardly rectifying potassium channel subunit, cause leber congenital amaurosis. Am J Hum Genet. 2011;89(1):183–90.
Orozco LD, et al. Integration of eQTL and a single-cell atlas in the Human Eye identifies causal genes for age-related Macular Degeneration. Cell Rep. 2020;30(4):1246–e12596.
Leach LL, Clegg DO. Concise Review: making stem cells retinal: methods for deriving retinal pigment epithelium and implications for patients with ocular disease. Stem Cells. 2015;33(8):2363–73.
Dehghan S, et al. Human-induced pluripotent stem cells-derived retinal pigmented epithelium, a new horizon for cells-based therapies for age-related macular degeneration. Stem Cell Res Ther. 2022;13(1):217.
Idelson M, et al. Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells. Cell Stem Cell. 2009;5(4):396–408.
Rouhani FJ, et al. Substantial somatic genomic variation and selection for BCOR mutations in human induced pluripotent stem cells. Nat Genet. 2022;54(9):1406–16.
Brandl C, et al. In-depth characterisation of Retinal Pigment Epithelium (RPE) cells derived from human induced pluripotent stem cells (hiPSC). Neuromolecular Med. 2014;16(3):551–64.
Liao JL, et al. Molecular signature of primary retinal pigment epithelium and stem-cell-derived RPE cells. Hum Mol Genet. 2010;19(21):4229–38.
Dalvi S, Galloway CA, Singh R. Pluripotent stem cells to Model degenerative retinal diseases: the RPE Perspective. Adv Exp Med Biol. 2019;1186:1–31.
Maeda T, et al. Strategies of pluripotent stem cell-based therapy for retinal degeneration: update and challenges. Trends Mol Med. 2022;28(5):388–404.
Oliveira CR, et al. Human stem cell-derived retinal pigment epithelial cells as a model for drug screening and pre-clinical assays compared to ARPE-19 cell line. Int J Stem Cells. 2021;14(1):74–84.
Pattnaik BR, et al. A novel KCNJ13 nonsense mutation and loss of Kir7.1 Channel function causes Leber congenital amaurosis (LCA16). Hum Mutat. 2015;36(7):720–7.
Xu P, et al. Biallelic CLCN2 mutations cause retinal degeneration by impairing retinal pigment epithelium phagocytosis and chloride channel function. Hum Genet. 2023;142(4):577–93.
Schwartz SD, et al. Subretinal transplantation of embryonic stem cell-derived retinal pigment epithelium for the Treatment of Macular Degeneration: an Assessment at 4 years. Invest Ophthalmol Vis Sci. 2016;57(5):ORSFc1–9.
Mandai M, et al. Autologous Induced stem-cell-derived retinal cells for Macular Degeneration. N Engl J Med. 2017;376(11):1038–46.
Wulff H, Castle NA, Pardo LA. Voltage-gated potassium channels as therapeutic targets. Nat Rev Drug Discov. 2009;8(12):982–1001.
Zhang X, Yang D, Hughes BA. KCNQ5/K(v)7.5 potassium channel expression and subcellular localization in primate retinal pigment epithelium and neural retina. Am J Physiol Cell Physiol. 2011;301(5):C1017–26.
Zhang X, Hughes BA. KCNQ and KCNE potassium channel subunit expression in bovine retinal pigment epithelium. Exp Eye Res. 2013;116:424–32.
Korkka I, Skottman H, Nymark S. Heterogeneity of Potassium channels in human embryonic stem cell-derived retinal pigment epithelium. Stem Cells Transl Med. 2022;11(7):753–66.
Wen R, Lui GM, Steinberg RH. Whole-cell K + currents in fresh and cultured cells of the human and monkey retinal pigment epithelium. J Physiol. 1993;465:121–47.
Mamaeva D, et al. Novel roles for voltage-gated T-type ca(2+) and ClC-2 channels in phagocytosis and angiogenic factor balance identified in human iPSC-derived RPE. FASEB J. 2021;35(4):e21406.
Yang D, Zhang X, Hughes BA. Expression of inwardly rectifying potassium channel subunits in native human retinal pigment epithelium. Exp Eye Res. 2008;87(3):176–83.
Hughes BA, Takahira M. Inwardly rectifying K + currents in isolated human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci. 1996;37(6):1125–39.
Yuan Y, Shimura M, Hughes BA. Regulation of inwardly rectifying K + channels in retinal pigment epithelial cells by intracellular pH. J Physiol. 2003;549(Pt 2):429–38.
Pattnaik BR, Hughes BA. Regulation of Kir channels in bovine retinal pigment epithelial cells by phosphatidylinositol 4,5-bisphosphate. Am J Physiol Cell Physiol. 2009;297(4):C1001–11.
Kumar M, Pattnaik BR. Focus on Kir7.1: physiology and channelopathy. Channels (Austin). 2014;8(6):488–95.
Yang D, et al. Expression and localization of the inwardly rectifying potassium channel Kir7.1 in native bovine retinal pigment epithelium. Invest Ophthalmol Vis Sci. 2003;44(7):3178–85.
Toms M, et al. Phagosomal and mitochondrial alterations in RPE may contribute to KCNJ13 retinopathy. Sci Rep. 2019;9(1):3793.
Yang D, et al. Expression of Kir7.1 and a novel Kir7.1 splice variant in native human retinal pigment epithelium. Exp Eye Res. 2008;86(1):81–91.
Kanzaki Y, et al. KCNJ13 gene deletion impairs cell alignment and phagocytosis in Retinal Pigment Epithelium Derived from Human-Induced pluripotent stem cells. Invest Ophthalmol Vis Sci. 2020;61(5):38.
Hejtmancik JF, et al. Mutations in KCNJ13 cause autosomal-dominant snowflake vitreoretinal degeneration. Am J Hum Genet. 2008;82(1):174–80.
Perez-Roustit S, et al. Leber congenital amaurosis with large retinal pigment clumps caused by compound heterozygous mutations in Kcnj13. Retin Cases Brief Rep. 2017;11(3):221–6.
Zhong H, et al. CRISPR-engineered mosaicism rapidly reveals that loss of Kcnj13 function in mice mimics human disease phenotypes. Sci Rep. 2015;5:8366.
Villanueva S, et al. Cleft palate, Moderate Lung Developmental Retardation and early postnatal lethality in mice deficient in the Kir7.1 inwardly rectifying K + Channel. PLoS ONE. 2015;10(9):e0139284.
Vera E, et al. Normal vision and development in mice with low functional expression of Kir7.1 in heterozygosis for a blindness-producing mutation inactivating the channel. Am J Physiol Cell Physiol. 2024;326(4):C1178–92.
Roman D, et al. Conditional loss of Kcnj13 in the retinal pigment epithelium causes photoreceptor degeneration. Exp Eye Res. 2018;176:219–26.
Ying G, et al. The small GTPase RAB28 is required for phagocytosis of cone outer segments by the murine retinal pigmented epithelium. J Biol Chem. 2018;293(45):17546–58.
Kanzaki Y, et al. Protrusion of KCNJ13 gene knockout retinal pigment epithelium due to oxidative stress-Induced cell death. Invest Ophthalmol Vis Sci. 2022;63(12):29.
Kabra M et al. Nonviral base editing of KCNJ13 mutation preserves vision in a model of inherited retinal channelopathy. J Clin Invest, 2023. 133(19).
Keckeis S, et al. Anoctamin2 (TMEM16B) forms the ca(2+)-activated Cl(-) channel in the retinal pigment epithelium. Exp Eye Res. 2017;154:139–50.
Schreiber R, Kunzelmann K. Expression of anoctamins in retinal pigment epithelium (RPE). Pflugers Arch. 2016;468(11–12):1921–9.
Hsu KS, et al. CLIC4 regulates late endosomal trafficking and matrix degradation activity of MMP14 at focal adhesions in RPE cells. Sci Rep. 2019;9(1):12247.
Liu J, et al. Cystic fibrosis transmembrane conductance regulator contributes to reacidification of alkalinized lysosomes in RPE cells. Am J Physiol Cell Physiol. 2012;303(2):C160–9.
Wimmers S, Karl MO, Strauss O. Ion channels in the RPE. Prog Retin Eye Res. 2007;26(3):263–301.
Marmorstein AD, et al. Bestrophin, the product of the best vitelliform macular dystrophy gene (VMD2), localizes to the basolateral plasma membrane of the retinal pigment epithelium. Proc Natl Acad Sci U S A. 2000;97(23):12758–63.
Barro Soria R, et al. Bestrophin-1 enables Ca2+-activated Cl- conductance in epithelia. J Biol Chem. 2009;284(43):29405–12.
Yang T, et al. Structure and selectivity in bestrophin ion channels. Science. 2014;346(6207):355–9.
Giblin JP, et al. Ion channels in the Eye: involvement in ocular pathologies. Adv Protein Chem Struct Biol. 2016;104:157–231.
Johnson AA, et al. Bestrophin 1 and retinal disease. Prog Retin Eye Res. 2017;58:45–69.
Habibi I et al. Clinical and genetic findings of autosomal recessive bestrophinopathy (ARB). Genes (Basel), 2019. 10(12).
Dalvin LA, et al. Retinitis pigmentosa associated with a mutation in BEST1. Am J Ophthalmol Case Rep. 2016;2:11–7.
Singh Grewal S, Smith JJ, Carr AF. Bestrophinopathies: perspectives on clinical disease, Bestrophin-1 function and developing therapies. Ther Adv Ophthalmol. 2021;13:2515841421997191.
Boulanger-Scemama E, et al. AUTOSOMAL DOMINANT VITREORETINOCHOROIDOPATHY: when Molecular Genetic Testing helps clinical diagnosis. Retina. 2019;39(5):867–78.
Guziewicz KE, et al. Bestrophin gene mutations cause canine multifocal retinopathy: a novel animal model for best disease. Invest Ophthalmol Vis Sci. 2007;48(5):1959–67.
Zangerl B, et al. Assessment of canine BEST1 variations identifies new mutations and establishes an independent bestrophinopathy model (cmr3). Mol Vis. 2010;16:2791–804.
Hoffmann I, et al. Canine multifocal retinopathy in the Australian shepherd: a case report. Vet Ophthalmol. 2012;15(Suppl 2):134–8.
Marmorstein LY, et al. The light peak of the electroretinogram is dependent on voltage-gated calcium channels and antagonized by bestrophin (best-1). J Gen Physiol. 2006;127(5):577–89.
Milenkovic A, et al. Bestrophin 1 is indispensable for volume regulation in human retinal pigment epithelium cells. Proc Natl Acad Sci U S A. 2015;112(20):E2630–9.
Zhang Y, et al. Suppression of Ca2 + signaling in a mouse model of best disease. Hum Mol Genet. 2010;19(6):1108–18.
Milenkovic A et al. The Y227N mutation affects bestrophin-1 protein stability and impairs sperm function in a mouse model of best vitelliform macular dystrophy. Biol Open, 2019. 8(7).
Carter DA, et al. Mislocalisation of BEST1 in iPSC-derived retinal pigment epithelial cells from a family with autosomal dominant vitreoretinochoroidopathy (ADVIRC). Sci Rep. 2016;6:33792.
Marmorstein AD, et al. Mutant Best1 expression and impaired phagocytosis in an iPSC model of autosomal recessive Bestrophinopathy. Sci Rep. 2018;8(1):4487.
Nachtigal AL et al. Mutation-dependent pathomechanisms determine the phenotype in the bestrophinopathies. Int J Mol Sci, 2020. 21(5).
Moshfegh Y, et al. BESTROPHIN1 mutations cause defective chloride conductance in patient stem cell-derived RPE. Hum Mol Genet. 2016;25(13):2672–80.
Ji C, et al. Investigation and restoration of BEST1 activity in patient-derived RPEs with Dominant mutations. Sci Rep. 2019;9(1):19026.
Singh R, et al. iPS cell modeling of best disease: insights into the pathophysiology of an inherited macular degeneration. Hum Mol Genet. 2013;22(3):593–607.
Singh R, et al. Pharmacological modulation of photoreceptor outer segment degradation in a human iPS cell model of inherited Macular Degeneration. Mol Ther. 2015;23(11):1700–11.
Lee JH, Oh JO, Lee CS. Induc Pluripotent Stem Cell Model Best Disease Autosomal Recessive Bestrophinopathy Yonsei Med J. 2020;61(9):816–25.
Li Y et al. Patient-specific mutations impair BESTROPHIN1’s essential role in mediating Ca(2+)-dependent Cl(-) currents in human RPE. Elife, 2017. 6.
Johnson AA, et al. Autosomal recessive bestrophinopathy is not Associated with the loss of Bestrophin-1 Anion Channel Function in a patient with a Novel BEST1 mutation. Invest Ophthalmol Vis Sci. 2015;56(8):4619–30.
Zhao Q et al. Distinct expression requirements and rescue strategies for BEST1 loss- and gain-of-function mutations. Elife, 2021. 10.
Amato A, et al. Gene therapy in bestrophinopathies: insights from preclinical studies in preparation for clinical trials. Saudi J Ophthalmol. 2023;37(4):287–95.
Ji C, et al. Dual ca(2+)-dependent gates in human Bestrophin1 underlie disease-causing mechanisms of gain-of-function mutations. Commun Biol. 2019;2:240.
Navines-Ferrer A et al. Impaired Bestrophin Channel activity in an iPSC-RPE model of best vitelliform macular dystrophy (BVMD) from an early onset patient carrying the P77S Dominant Mutation. Int J Mol Sci, 2022. 23(13).
Cordes M, et al. Inhibition of ca(2+) channel surface expression by mutant bestrophin-1 in RPE cells. FASEB J. 2020;34(3):4055–71.
Duncker T, et al. Quantitative fundus autofluorescence and optical coherence tomography in best vitelliform macular dystrophy. Invest Ophthalmol Vis Sci. 2014;55(3):1471–82.
Lin TC, et al. Nanomedicine-based Curcumin Approach Improved ROS damage in best dystrophy-specific Induced Pluripotent Stem cells. Cell Transpl. 2019;28(11):1345–57.
Liu J, et al. Small Molecules Restore Bestrophin 1 expression and function of both Dominant and recessive bestrophinopathies in patient-derived retinal pigment epithelium. Invest Ophthalmol Vis Sci. 2020;61(5):28.
Elverson K et al. Tadalafil rescues the p.M325T mutant of Best1 Chloride Channel. Molecules, 2023. 28(8).
Steyer B, et al. Scarless Genome Editing of Human pluripotent stem cells via transient puromycin selection. Stem Cell Rep. 2018;10(2):642–54.
Thiemann A, et al. A chloride channel widely expressed in epithelial and non-epithelial cells. Nature. 1992;356(6364):57–60.
Wang H, et al. Research and progress on ClC2 (review). Mol Med Rep. 2017;16(1):11–22.
Jentsch TJ, Pusch M. CLC Chloride Channels and transporters: structure, function, physiology, and Disease. Physiol Rev. 2018;98(3):1493–590.
Depienne C, et al. Brain white matter oedema due to ClC-2 chloride channel deficiency: an observational analytical study. Lancet Neurol. 2013;12(7):659–68.
Min R, et al. In: Adam MP, et al. editors. CLCN2-Related Leukoencephalopathy, in GeneReviews((R)). Seattle (WA); 1993.
Niemeyer MI, et al. No evidence for a role of CLCN2 variants in idiopathic generalized epilepsy. Nat Genet. 2010;42(1):3.
Kleefuss-Lie A, et al. CLCN2 variants in idiopathic generalized epilepsy. Nat Genet. 2009;41(9):954–5.
Fernandes-Rosa FL, et al. A gain-of-function mutation in the CLCN2 chloride channel gene causes primary aldosteronism. Nat Genet. 2018;50(3):355–61.
Scholl UI, et al. CLCN2 chloride channel mutations in familial hyperaldosteronism type II. Nat Genet. 2018;50(3):349–54.
Di Bella D, et al. Subclinical leukodystrophy and infertility in a man with a novel homozygous CLCN2 mutation. Neurology. 2014;83(13):1217–8.
Giorgio E, et al. A novel homozygous change of CLCN2 (p.His590Pro) is associated with a subclinical form of leukoencephalopathy with ataxia (LKPAT). J Neurol Neurosurg Psychiatry. 2017;88(10):894–6.
Guo Z, et al. CLCN2-related leukoencephalopathy: a case report and review of the literature. BMC Neurol. 2019;19(1):156.
Cheng Y, et al. Case report: a frameshift mutation in CLCN2-related leukoencephalopathy and retinopathy. Front Genet. 2023;14:1278961.
Edwards MM, et al. Photoreceptor degeneration, azoospermia, leukoencephalopathy, and abnormal RPE cell function in mice expressing an early stop mutation in CLCN2. Invest Ophthalmol Vis Sci. 2010;51(6):3264–72.
Krebs MP, et al. Mouse models of human ocular disease for translational research. PLoS ONE. 2017;12(8):e0183837.
Nehrke K, et al. Loss of hyperpolarization-activated Cl(-) current in salivary acinar cells from Clcn2 knockout mice. J Biol Chem. 2002;277(26):23604–11.
Bosl MR, et al. Male germ cells and photoreceptors, both dependent on close cell-cell interactions, degenerate upon ClC-2 Cl(-) channel disruption. EMBO J. 2001;20(6):1289–99.
Goppner C, et al. Cellular basis of ClC-2 Cl(-) channel-related brain and testis pathologies. J Biol Chem. 2021;296:100074.
Hanke-Gogokhia C, et al. Apical CLC-2 in retinal pigment epithelium is crucial for survival of the outer retina. FASEB J. 2021;35(7):e21689.
Korkka I, et al. Characterization of Chloride channels in human embryonic stem cell derived retinal pigment epithelium. Singapore: Springer Singapore; 2018.
Hazim RA, Williams DS. Microtubule Motor Transport of Organelles in a Specialized Epithelium: the RPE. Front Cell Dev Biol. 2022;10:852468.
Rieu Q et al. Pleiotropic Roles of Scavenger Receptors in circadian retinal phagocytosis: a new function for lysosomal SR-B2/LIMP-2 at the RPE Cell Surface. Int J Mol Sci, 2022. 23(7).
Kono R, Ikegaya Y, Koyama R. Phagocytic glial cells in Brain Homeostasis. Cells, 2021. 10(6).
Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2018;18(4):225–42.
Panneerdoss S, et al. Cross-talk between mir-471-5p and autophagy component proteins regulates LC3-associated phagocytosis (LAP) of apoptotic germ cells. Nat Commun. 2017;8(1):598.
Nighot PK, Leung L, Ma TY. Chloride channel ClC- 2 enhances intestinal epithelial tight junction barrier function via regulation of caveolin-1 and caveolar trafficking of occludin. Exp Cell Res. 2017;352(1):113–22.
Jin Y, et al. Knockout of ClC-2 reveals critical functions of adherens junctions in colonic homeostasis and tumorigenicity. Am J Physiol Gastrointest Liver Physiol. 2018;315(6):G966–79.
Blaug S, et al. Retinal pigment epithelial function: a role for CFTR? Doc Ophthalmol. 2003;106(1):43–50.
Chuang JZ, et al. Retinal pigment epithelium-specific CLIC4 mutant is a mouse model of dry age-related macular degeneration. Nat Commun. 2022;13(1):374.
Cao X, Pattnaik BR, Hughes BA. Mouse retinal pigment epithelial cells exhibit a thiocyanate-selective conductance. Am J Physiol Cell Physiol. 2018;315(4):C457–73.
Hanssens LS, Duchateau J, Casimir GJ. CFTR Protein: Not Just Chloride Channel? Cells, 2021. 10(11).
Fico G et al. Characterization of Chloride Channels in Human Embryonic Stem Cell Derived Retinal Pigment Epithelium, in IFMBE Proceedings. 2018. pp. 1089–1090.
Li R, et al. CNTF mediates neurotrophic factor secretion and fluid absorption in human retinal pigment epithelium. PLoS ONE. 2011;6(9):e23148.
Wu J, Marmorstein AD, Peachey NS. Functional abnormalities in the retinal pigment epithelium of CFTR mutant mice. Exp Eye Res. 2006;83(2):424–8.
Duncan RR, et al. Rat brain p64H1, expression of a new member of the p64 chloride channel protein family in endoplasmic reticulum. J Biol Chem. 1997;272(38):23880–6.
Argenzio E, Moolenaar WH. Emerging biological roles of Cl- intracellular channel proteins. J Cell Sci. 2016;129(22):4165–74.
Chuang JZ, Chou SY, Sung CH. Chloride intracellular channel 4 is critical for the epithelial morphogenesis of RPE cells and retinal attachment. Mol Biol Cell. 2010;21(17):3017–28.
Yu HG, et al. Increased choroidal neovascularization following laser induction in mice lacking lysyl oxidase-like 1. Invest Ophthalmol Vis Sci. 2008;49(6):2599–605.
Cheng J, Hao X, Zhang Z. Risk of macular degeneration affected by polymorphisms in Matrix metalloproteinase-2: a case-control study in Chinese Han population. Med (Baltim). 2017;96(47):e8190.
Krueger B, et al. Four subunits (alphabetagammadelta) of the epithelial sodium channel (ENaC) are expressed in the human eye in various locations. Invest Ophthalmol Vis Sci. 2012;53(2):596–604.
Berendt FJ, Park KS, Trimmer JS. Multisite phosphorylation of voltage-gated sodium channel alpha subunits from rat brain. J Proteome Res. 2010;9(4):1976–84.
Nishino A, Okamura Y. Evolutionary history of Voltage-gated Sodium channels. Handb Exp Pharmacol. 2018;246:3–32.
Kwong K, Carr MJ. Voltage-gated sodium channels. Curr Opin Pharmacol. 2015;22:131–9.
Reichhart N, Strauss O. Ion channels and transporters of the retinal pigment epithelium. Exp Eye Res. 2014;126:27–37.
Lidgerwood GE, et al. Defined medium conditions for the induction and expansion of human pluripotent stem cell-derived retinal pigment epithelium. Stem Cell Rev Rep. 2016;12(2):179–88.
Ignatova I, Frolov R, Nymark S. The retinal pigment epithelium displays electrical excitability and lateral signal spreading. BMC Biol. 2023;21(1):84.
Karema-Jokinen V, et al. Crosstalk of protein clearance, inflammasome, and ca(2+) channels in retinal pigment epithelium derived from age-related macular degeneration patients. J Biol Chem. 2023;299(6):104770.
Wimmers S, et al. Expression profile of voltage-dependent Ca2 + channel subunits in the human retinal pigment epithelium. Graefes Arch Clin Exp Ophthalmol. 2008;246(5):685–92.
Rosenthal R, Strauss O. Ca2+-channels in the RPE. Adv Exp Med Biol. 2002;514:225–35.
Reichhart N, et al. Rab27a GTPase modulates L-type Ca2 + channel function via interaction with the II-III linker of CaV1.3 subunit. Cell Signal. 2015;27(11):2231–40.
Rosenthal R, et al. Ca2 + channels in retinal pigment epithelial cells regulate vascular endothelial growth factor secretion rates in health and disease. Mol Vis. 2007;13:443–56.
Muller C, et al. CaV1.3 L-type channels, maxiK ca(2+)-dependent K(+) channels and bestrophin-1 regulate rhythmic photoreceptor outer segment phagocytosis by retinal pigment epithelial cells. Cell Signal. 2014;26(5):968–78.
Etheredge JA, et al. Functional compensation by other voltage-gated Ca2 + channels in mouse basal forebrain neurons with ca(V)2.1 mutations. Brain Res. 2007;1140:105–19.
Krizaj D, Cordeiro S, Strauss O. Retinal TRP channels: cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res. 2023;92:101114.
Tan LX, et al. Optineurin tunes outside-in signaling to regulate lysosome biogenesis and phagocytic clearance in the retina. Curr Biol. 2023;33(18):3805–e38207.
Vergarajauregui S, Martina JA, Puertollano R. LAPTMs regulate lysosomal function and interact with mucolipin 1: new clues for understanding mucolipidosis type IV. J Cell Sci. 2011;124(Pt 3):459–68.
Di Paola S, Scotto-Rosato A, Medina DL. TRPML1: the ca((2+))retaker of the lysosome. Cell Calcium. 2018;69:112–21.
Bargal R, et al. Identification of the gene causing mucolipidosis type IV. Nat Genet. 2000;26(1):118–23.
Grishchuk Y, et al. Retinal dystrophy and Optic nerve Pathology in the mouse model of mucolipidosis IV. Am J Pathol. 2016;186(1):199–209.
Gomez NM, et al. Robust lysosomal calcium signaling through channel TRPML1 is impaired by lysosomal lipid accumulation. FASEB J. 2018;32(2):782–94.
Beckel JM, et al. Stimulation of TLR3 triggers release of lysosomal ATP in astrocytes and epithelial cells that requires TRPML1 channels. Sci Rep. 2018;8(1):5726.
Kiss S, et al. Long-term safety evaluation of continuous intraocular delivery of Aflibercept by the Intravitreal Gene Therapy Candidate ADVM-022 in Nonhuman Primates. Transl Vis Sci Technol. 2021;10(1):34.
Schwartz SD, et al. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet. 2012;379(9817):713–20.
da Cruz L, et al. Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol. 2018;36(4):328–37.
Humayun MS, et al. Long-term follow-up of a phase 1/2a clinical trial of a stem cell-derived Bioengineered Retinal Pigment Epithelium Implant for Geographic Atrophy. Ophthalmology. 2024;131(6):682–91.
da Cruz L et al. The fate of RPE cells following hESC-RPE Patch Transplantation in Haemorrhagic Wet AMD: pigmentation, extension of pigmentation, thickness of Transplant, Assessment for Proliferation and visual Function-A 5 year-follow up. Diagnostics (Basel), 2024. 14(10).
Gupta S, et al. Progress in stem cells-based replacement therapy for retinal pigment epithelium: in Vitro differentiation to in vivo delivery. Stem Cells Transl Med. 2023;12(8):536–52.
Xu Z, et al. A single-cell transcriptome atlas of the human retinal pigment epithelium. Front Cell Dev Biol. 2021;9:802457.
Ortolan D, et al. Single-cell-resolution map of human retinal pigment epithelium helps discover subpopulations with differential disease sensitivity. Proc Natl Acad Sci U S A. 2022;119(19):e2117553119.
Lee JR, Jeong KW. NMDA receptor antagonists degrade lipofuscin via Autophagy in Human Retinal pigment epithelial cells. Med (Kaunas), 2022. 58(8).
Liu S, et al. Self-formation of RPE spheroids facilitates Enrichment and expansion of hiPSC-Derived RPE generated on retinal organoid induction platform. Invest Ophthalmol Vis Sci. 2018;59(13):5659–69.
Michelet F, et al. Rapid generation of purified human RPE from pluripotent stem cells using 2D cultures and lipoprotein uptake-based sorting. Stem Cell Res Ther. 2020;11(1):47.
Regent F, et al. Automation of human pluripotent stem cell differentiation toward retinal pigment epithelial cells for large-scale productions. Sci Rep. 2019;9(1):10646.
Rizzolo LJ, Nasonkin IO, Adelman RA. Retinal cell transplantation, Biomaterials, and in Vitro models for developing next-generation therapies of age-related Macular Degeneration. Stem Cells Transl Med. 2022;11(3):269–81.
Acknowledgements
The authors would like to thank all the reviewers who participated in the review and MJEditor (www.mjeditor.com) for its linguistic assistance during the preparation of this manuscript.
Funding
This work was supported by the National Key Research and Development Program of the Ministry of Science and Technology (2023YFC2506100), the National Natural Science Foundation of China (81970842, 82172957), A joint grant from Science & Technology Project of Guangzhou and Zhongshan Ophthalmic Center, Sun Yat-sen University (202201020312), The Science &Technology Project of Guangzhou (202102010288), China Postdoctoral Science Foundation (2023M744064, 2021M703705) and Guangdong Basic and Applied Basic Research Foundation (2022A1515110631).
Author information
Authors and Affiliations
Contributions
PX was responsible for drafting the manuscript, creating figures, and designing tables. WSZ, WJY, GFC, and GJG contributed to drafting and revising the manuscript. XFZ designed and revised the manuscript and provided financial support for publication. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Xu, P., Zou, W., Yin, W. et al. Ion channels research in hPSC-RPE cells: bridging benchwork to clinical applications. J Transl Med 22, 1073 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05769-5
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-024-05769-5