325
Views
17
CrossRef citations to date
0
Altmetric
Review

Stem cell therapies for age-related macular degeneration: the past, present, and future

, &
Pages 255-264 | Published online: 14 Jan 2015

Abstract

In the developed world, age-related macular degeneration (AMD) is one of the major causes of irreversible blindness in the elderly. Although management of neovascular AMD (wet AMD) has dramatically progressed, there is still no effective treatment for nonneovascular AMD (dry AMD), which is characterized by retinal pigment epithelial (RPE) cell death (or dysfunction) and microenvironmental disruption in the retina. Therefore, RPE replacement and microenvironmental regulation represent viable treatments for dry AMD. Recent advances in cell biology have demonstrated that RPE cells can be easily generated from several cell types (pluripotent stem cells, multipotent stem cells, or even somatic cells) by spontaneous differentiation, coculturing, defined factors or cell reprogramming, respectively. Additionally, in vivo studies also showed that the restoration of visual function could be obtained by transplanting functional RPE cells into the subretinal space of recipient. More importantly, clinical trials approved by the US government have shown promising prospects in RPE transplantation. However, key issues such as implantation techniques, immune rejection, and xeno-free techniques are still needed to be further investigated. This review will summarize recent advances in cell transplantation for dry AMD. The obstacles and prospects in this field will also be discussed.

Background

In the Western world, age-related macular degeneration (AMD) is one of the leading causes of blindness in the elderly. The incidence rate of AMD has continued to increase in the past decades.Citation1Citation4 According to the presence or absence of choroidal neovascularization, advanced AMD can be generally classified into two types: dry AMD and wet AMD. Wet AMD could be controlled by drugs that target the vascular endothelial growth factor (VEGF), photodynamic therapy, laser photocoagulation, and vitrectomy at different phases. Dry AMD, which is primarily attributed to the accumulation of reactive oxygen species and lipid peroxide, can evoke chronic inflammations in the retina and lead to apoptosis of the retinal pigment epithelial (RPE) cells, and finally damages the photoreceptors.Citation5 Currently, no treatments can reverse dry AMD, regardless of the fact that dietary supplementation with defined vitamins and antioxidants has been shown to alleviate progression.Citation6 Therefore, RPE replacement and retinal microenvironmental regulation represent potential new approaches for dry AMD.

Functional RPE cells could be generated from stem cells or somatic cells by spontaneous differentiation,Citation7Citation16 coculturing,Citation17 defined factors,Citation18Citation22 or cell reprogramming.Citation23 Source of RPE cells for transplantation seems to be unlimited. More importantly, a clinical trial approved by the US government has shown promising prospects in RPE transplantation.Citation24 However, xeno-free techniques,Citation11,Citation12 implantation techniques, immune rejection,Citation25Citation27 and the safety issues are still under debate.

In addition, mesenchymal stem cells (MSCs) have various biological effects,Citation28 such as immunoregulation, antiapoptosis of neurons, and neurotrophin secretion. In vivo studies also have suggested that MSCs could recover and regulate the retinal microenvironment in different models of retinal degeneration.Citation29,Citation30 Moreover, MSCs are also ideal vehicles in cell engineering. Gene-modified MSCs always have specific functions and could be utilized in AMD treatments.Citation31Citation34

This review will focus on the following aspects: 1) RPE transplantation and 2) stem cell-based retinal microenvironmental regulation.

RPE transplantation

Healthy and vigorous RPE cells are ideal donors for transplantation, and pre-AMD is a viable therapeutic target. According to the cell source, they could be divided into 1) autologous RPE cells, 2) stem cell-derived RPE cells, and 3) reprogrammed RPE cells.

Autologous RPE cells

As the diseased RPE is a major component of dry AMD, several attempts have been made to replace the aged RPE cells located at the macula. Macular translocation surgery is conducted by the detachment and rotation of neural retina from the diseased macular RPE layer to another healthy place.Citation35Citation37 After up to 5 years of follow-up, three Snellen lines of improvement in best corrected visual acuity were obtained in some patients.Citation38Citation40 However, high complication rates were noticed, such as macular edema, retinal detachment, double vision, and cataract formation.Citation38Citation40 Nonetheless, successes in macular translocation demonstrated that 1) healthy RPE cells were located in the diseased retina and 2) these healthy RPE cells could restore the visual function in AMD patients.

Thereafter, autologous RPE transplantation as an alternative surgical approach was widely studied. It is accomplished by collecting healthy RPE cells in the peripheral retina and transplanting them into the subretinal space at the diseased macula.Citation41Citation45 The clinical outcomes are similar to those of the macular translocation: maintenance or slight elevations in visual acuity were reported in several trials after 3 or 4 years of follow-up.Citation41Citation44 Although autologous RPE transplantation has a relatively low rate of complication when compared with macular translocation, there are some remarkable drawbacks: 1) The initial harvesting of RPE cells from patients increases the length of the surgical procedure and the risk of postsurgery complications, such as cataract formation and retinal detachment. 2) No evidence could demonstrate that the transplanted RPE cells in suspension can first attach to the diseased Bruch’s membrane and form the desired monolayer which is required for optimal RPE function. In contrast, these cells always clump into rosettesCitation46 or undergo anoikis,Citation47 a form of apoptosis specific to anchorage-dependent cells that are dissociated from their usual extracellular matrix. 3) The cells being harvested are the same age as the cells they are designed to be replaced. 4) Autologous RPE transplantation requires more than 60,000 viable RPE cells. It is quite difficult to collect enough cells to repopulate the entire macula adequately.

Stem cell-derived RPE cells

RPE cells from the patients might be insufficient for transplantation, and highly efficient protocols for generating functional RPE cells are eagerly required. Stem cells are able to differentiate into several cell types as well as self-renew. According to their potential, they can be generally classified into pluripotent stem cells (embryonic stem cells [ESCs] and induced pluripotent stem cells [iPSCs]) and multipotent stem cells (neural stem cells, MSCs, and so on). Recent studies have revealed that 1) functional RPE cells could be differentiated from pluripotent stem cells or multipotent stem cells by defined protocols and 2) visual function could be restored in vivo by transplantation of stem cell-derived RPE cells.

ESC-derived RPE cells

ESCs have extensive abilities to differentiate into all three germ layers. In the past decade, with the development of cell sciences, ESCs manifest extremely attractive prospects in the treatment of degenerative diseases. Several defined protocols were conducted to generate mature RPE cells from ESCs.

Spontaneous differentiation

Adherent culture

In natural conditions, ESCs can spontaneously differentiate into RPE-like cells by adherent culturing. This was originally reported by Kawasaki et al.Citation7 They found that about 8%±4% of pigmented cells could be generated from primate ESCs by coculturing with PA6 stromal cells. The ESC-derived RPE cells were hexagonal and contained significant amounts of pigment. They also expressed the mature markers of RPE cell: ZO-1, RPE65, CRALBP, and MerTK. Electron microscopy revealed that these cells had extensive microvilli and were able to phagocytose latex beads. After transplanting into the subretinal space of RCS (Royal College of Surgeons, London, UK) rats (a well-known model of RPE degeneration, which has a mutation in MerTK, characterized by loss of phagocytic function of RPE cells), the grafted RPE cells increased the survival of host photoreceptors. Histologic analyses and behavioral tests further confirmed this.Citation8 This protocol has multiple advantages: 1) the techniques were relatively simple, and ESCs were only seeded onto the PA6 stromal cells feeder to form colonies; 2) neural differentiation of ESCs is efficient and speedy; and 3) no exogenous reagent was used. But harvested cells could be contaminated by PA6 stromal cells.

Human RPE cells also could be differentiated from human ESCs (hESCs) by similar ways: hESCs were seeded on an inactivated feeder and allowed to overgrow until confluent (approximately 2 weeks). Then, the basic fibroblast growth factor was removed from the medium, and the cells were allowed to differentiate spontaneously. After 4–5 weeks, pigmented foci could be observed. When these cells were transplanted into the subretinal space of RCS rats, the cells displayed polarity and integrated well into the host retina. More importantly, these cells showed phagocytic functions. Improvement in visual performance was 100% over untreated controls (spatial acuity was approximately 70% that of normal nondystrophic rats). In the safety evaluation, teratoma formation and other pathological changes were not observed under immunosupression.Citation9,Citation10 Although the efficiency is relatively low, a significant advantage of this protocol is that no additional reagents (such as Wnt or nodal inhibitors) were supplemented. This protocol mimics the natural generation of RPE cells and avoids the potential contaminations from recombinant proteins or small molecules which were used in other protocols.

However, most of the published protocols used mouse embryonic fibroblast cells as the feeder layer for hESCs and human induced pluripotent stem cells (hiPSCs). Xenoproducts used in the differentiation processes pose further challenges, because animal-derived components may carry factors such as sialic acid or Neu5Gc, causing unwanted immunogenicity of the cells,Citation48,Citation49 or even animal pathogens. Recently, Vaajasaari et alCitation11 and Zhang et alCitation12 reported the differentiation of functional RPE-like cells from several hESC lines and one hiPSC line in defined and xeno-free conditions, providing an important step toward a defined and xeno-free culture and differentiation process, enabling easy translation to clinical-quality cell production under Good Manufacturing Practice regulations.

Suspension culture

RPE cells could also be spontaneously differentiated from ESCs by embryonic formation. ESCs were seeded onto a petri dish in the absence of a differentiation antagonist to form embryonic bodies (EBs). Three-dimensional suspension aggregates can mimic embryonic development in vivo. To yield more cells of neuroectodermal lineage, the EBs are replated to a coated dish containing neural differentiation media (coated with extracellular matrix) for adherent culturing.Citation13,Citation14 Pigmented cells could be found thereafter. In 2011, Advanced Cell Technology (Santa Monica, CA, USA) performed Phase I/II clinical trials by using this protocol to elucidate the efficiencies of hESC-derived RPE transplantation on dry AMD and Stargardt’s disease (registration numbers NCT1345006 and NCT1344993).Citation50 Subsequently, Schwartz et alCitation24 published the preliminary results of their study, in which two patients (dry AMD and Stargardt’s disease, respectively) received subretinal transplantation of 5×104 induced RPE cells by vitrectomy. Efficiency evaluations: the cells survived after 4 months of follow-up. The best corrected visual acuities of both patients were slightly improved: 7-letter improvements were achieved in the AMD patient (from 21 to 28 letters) and 5-letter improvements were achieved for the patient with Stargardt’s disease (evaluated by the Early Treatment for Diabetic Retinopathy Study visual chart). Safety evaluations: no teratoma formation and immunologic rejection were noticed in both cases. The investigators also found that the phase of cell differentiation was directly associated with cellular attachment and survival: RPE cells with mild depigmentation have better proliferative and adherent abilities. Therefore, choosing donor cells at optimal stages is a crucial step for successful transplantation. Also, hESCs used for differentiation should not contain pathogenic genes, and RPE cell purification is an additional concern.

Subsequently, with the establishment of a three-dimensional culture system, Eiraku et alCitation51 reported that the optic cup and mature RPE layers could be spontaneously generated by a three-dimensional culture of mouse ESC aggregates. Zhu et alCitation52 demonstrated the utility of this epithelial culture approach by achieving a quantitative production of RPE cells from hESCs within 30 days. Direct transplantation of this RPE into a rat model of retinal degeneration without any selection or expansion of the cells results in the formation of a donor-derived RPE monolayer that rescues photoreceptor cells. The cyst method for neuroepithelial differentiation of pluripotent stem cells is not only of importance for RPE generation but will also be relevant to the production of other neuronal cell types and for reconstituting complex patterning events from three-dimensional neuroepithelia.

However, three-dimensional culturing is time-consuming and expensive. To enhance the efficiency of RPE generation, Cho et alCitation53 conducted a protocol which indicated that RPE cells could be obtained from spherical neural masses. The target cells showed polygonal-shaped epithelial monolayer, and electron microscopy revealed apical microvilli, pigment granules, and tight junctions. These cells also expressed molecular markers of RPE, including ZO-1, RPE65, and bestrophin. On functional evaluation, these cells showed phagocytosis of isolated photoreceptor outer segment (POS) and secretion of soluble factors such as pigment epithelium-derived factor (PEDF) and VEGF. This protocol has remarkable merits: 1) Spherical neural masses have the capability of expansion for long periods without loss of differentiation capability and 2) they are easy to store and thaw, and there is no need for feeder cells. Thus, it could be an efficient strategy for obtaining functional RPE cells for retinal regenerative therapy.

Directed differentiation

ESCs also can directly differentiate into RPE-like cells by supplementing with defined factors.Citation18,Citation19 Early studies using stepwise differentiation protocols were based on models of telencephalic cell derived from ESCs, and combined EB formation with subsequent culture of attached cells in media containing proteins which control specification of neuronal lineage (such as Dkk1, a Wnt antagonist; LeftyA, a nodal antagonist). In 2005, Ikeda et alCitation18 conducted a protocol by which retinal precursors could be directly differentiated from mouse ESCs by supplementing Dkk1 and LeftyA under serum-free, feeder-free conditions; 16% of the total cells could be differentiated into retinal precursor cells (Rax positive). After optimizing the protocols, the efficiency of differentiation has been greatly elevated.Citation19

In addition, insulin-like growth factor signaling pathways and transforming growth factor beta (TGFβ) signaling pathways (such as bone morphogenetic protein antagonists, nicotinamide, and Activin A) were also reported to play important roles in RPE differentiation. Using Noggin (a bone morphogenetic protein antagonist), Dkk1-1, and insulin-like growth factor 1, Lamba et alCitation54 found that up to 80% of the H1 line can be directed to the retinal progenitor fate, and express a gene expression profile similar to that of progenitors derived from human fetal retina. The most prominent benefit of this protocol is that high percentages of target cells were generated from hESCs within a short period. In another study, Idelson et alCitation55 revealed that nicotinamide (belonging to TGFβ superfamily), which presumably patterns RPE development during embryogenesis, promotes the differentiation of hESCs to neural and subsequently to RPE fate. The hESC-derived RPE cells exhibited a morphology, marker expression, and function similar to those of authentic RPE and restored retinal structure and function after transplantation in vivo. Activin A, a member of the TGFβ superfamily, is another critical factor in RPE differentiation. It was secreted by the extracellular mesenchyme during optic cup development. With the addition of Activin A, the yield of RPE cells increased.Citation55 Alternatively, Activin A may serve to maintain the differentiated RPE cell phenotype in culture.Citation56

Although protocols mentioned so far have become more efficient than the report in 2004, they are still a bit time-consuming and inefficient. In a recent study, Buchholz et alCitation57 found that supplementing with defined factors at specific times could yield approximately 80% of the cells to an RPE phenotype within 2 weeks. They also noticed that culturing with more non-RPE cells led to faster RPE pigmentation, suggesting that these cells may secrete factors that activate melanogenesis.

However, the defined factors in these protocols are all derived from animal cells or Escherichia coli, raising the possibility of infection or immune rejection due to cross-species contamination. By contrast, using chemical compounds offers several advantages, compared with the recombinant proteins: 1) the small molecules are chemicals, which are consistent between different lot numbers and manufacturers; 2) the cross-species contaminations and cross-reactions are easily avoided; and 3) the cost is relatively low, making this method applicable. In a serum-free and feeder-free floating aggregate culture, Osakada et alCitation58 found that ESCs and iPSCs could be efficiently differentiated into RPE cells by supplementing CKI-7 (a Wnt antagonist) and SB-431542 (a nodal antagonist). These cells displayed the characteristic morphology of mature RPE cells, protein markers, and phagocytic capacity. This method provides a solution to cross-species antigenic contamination in transplantation, and is also useful for in vitro modeling of development, disease, and drug screening. However, whether these effects are reversible and transient is largely unknown. More research is needed to evaluate the long-term biological effects.

iPSC-derived RPE cells

In recent years, the most breaking advance in cell biology is probably iPSCs, which was first reported by Takahashi and YamanakaCitation59 and Yu et al.Citation60 These cells reprogrammed by using Thomson factors or Yamanaka factors showed morphological characteristics and differentiation abilities (including iPSCs to RPE) similar to those of the ESCs. Studies by several groups have already demonstrated that human RPE cells could be generated from iPSCs by spontaneous differentiationCitation15,Citation16 or directed differentiation.Citation20Citation22,Citation61 The iPSC-derived RPE cells were morphologically similar to, and expressed numerous markers of, developing and mature RPE cells. Phagocytosis of isolated POS and secretion of soluble factors (PEDF and VEGF) were also mentioned by several groups.Citation15,Citation16,Citation20Citation22 Interestingly, Westenskow et alCitation62 developed a flow cytometry-based assay to compare the phagocytic function between ARPE-19, human fetal RPE, and two types of iPSCs-RPE. They found that highly differentiated iPSCs-RPE phagocytosed POS more efficiently than did native RPE. In vivo studies also suggested that transplantation of these cells could facilitate the maintenance of photoreceptors through phagocytosis of the POS in the model of RPE degeneration.Citation15,Citation63

Additionally, iPSCs could be generated by using less transcription factors, which would reduce the incidence of tumorigenesis. Krohne et alCitation63 found that 1-factor-iPSC-RPE significantly resembled native RPE cells not only on proteomics and untargeted metabolomic analyses but also on in vivo functional evaluations. They showed that 1-factor-iPSC-RPE mediates anatomical and functional rescue of photoreceptors after transplantation in an animal model of RPE degeneration. Moreover, iPSCs could also be derived from other somatic cells than fibroblasts, including RPE cells. Hu et alCitation64 reprogrammed primary RPE cells by using OCT4, SOX2, LIN28, and Nanog. The RPE-derived iPSCs exhibited morphologies, gene expressions, and teratoma formation similar to hESCs and other iPS cell lines. After spontaneous differentiation by the removal of fibroblast growth factor 2, the resultant RPE cells showed a marked preference for redifferentiation into RPE. They suggested that target cells retain a memory of their previous state of differentiation.

Despite the fact that most protocols for ESC differentiation are suitable for iPSCs, differentiation efficiencies between iPS cell lines vary. Hirami et alCitation61 suggested that under identical conditions (SFEB/DL), 201B7 and 253G1 cell lines could differentiate into RPE cells, whereas 201B6 cell lines could not. From the perspective of protein expression, 6 days after differentiation toward RPE cells, Rx+/Pax+ cells emerged in an mESC-derived pool of cells, whereas this emergence requires 15 days with cells derived from certain iPS cell lines.

iPSC-derived RPEs have several advantages. First, absence of ethical concerns is the biggest benefit for research. Second, patient-specific iPSCs might have minimal immunogenicity than ESCs or other originated RPE cells. Third, iPSC-derived RPEs could be considered as a well-established model for disease mimicking and drug screening.

However, shortcomings of using iPSC-derived RPEs for transplantation cannot be ignored: 1) cells derived from iPSCs have the potential ability of tumorigenesis, which would restrict their clinical applications; 2) generation of patient-specific iPSCs would be a costly and time-consuming course; and 3) patient-specific iPSCs might have genetic defects that contribute to the disease. Combining iPSC technology with gene therapy is a promising solution.Citation65

MSC-derived RPE cells

Although RPE cells are derived from the ectoderm, MSCs have the ability of cross-mesodermal differentiation. Huang et alCitation17 found that RPE-like cells could be obtained from MSCs by RPE conditional medium supplemented with POS. These cells have morphological features and phagocytic capabilities similar to those of the native RPE cells.

Moreover, studies have also indicated that retinal cells could be differentiated from MSCs and replace the damaged retinal cells under certain conditions.Citation66,Citation67 Gong et alCitation66 reported that MSC-originated RPE cells could be found in the sodium iodide-damaged retina after subretinal injection of MSCs for 5 days.

Retinal stem cell-derived RPE cells

The retinal stem cells (RSCs) are situated in the ciliary marginal zone (CMZ) in fish and amphibians. The CMZ can continuously generate new neurons after retinal injury. Despite the fact that the mature retina in mammalians lacks regenerative ability, Tropepe et alCitation68 noticed that CMZ cells are capable of proliferating and differentiating into retinal cells (rods, bipolar cells, and glial cells) in mature mice. By isolating RSCs and supplementing with linoleic acid, selenite, insulin, transferrin, thyroxin, and other factors into the medium, Aruta et alCitation69 successfully differentiated RSCs into polarized and phagocytotic RPE-like cells. Similar to the MSC-derived RPE cells described by Huang et alCitation17 no studies were conducted to evaluate the function and safety of induced RPE cells in vivo.

However, the existence of mammalian RSCs is still under debate. Cicero et alCitation70 speculated that the so-called RSCs are ciliary epithelial cells. Their study showed that no significant differences in molecular, cellular, and morphological characteristics were observed between RSCs and ciliary epithelial cells. They suggested that ciliary epithelial cells can form colony spheres, undergo self-renewal, and express precursor markers.

In addition, Müller cells were once considered as retinal stem cells. Bernardos et alCitation71 reported that Müller cells could express Pax6 and Crx at a low level in zebra fish. Song et alCitation72 found that Atoh7 could promote the transformation of Müller cells into retinal ganglion cells. However, Müller cells originate from neural retinal precursors and mature at the last stages of retinogenesis, and RPE precursors, and neural retinal precursors divided during early embryonic development (neural retinal cells develop in the following order: retinal ganglion cells, cone cells, amacrine cells, horizontal cells, rod cells, bipolar cells, and Müller cells). Therefore, direct transformation of Müller cells into RPE will be extremely difficult.

Reprogrammed RPE cells (somatic cell-derived RPE cells)

With the development of cell biology, direct cell reprogramming shows a promising prospect in the generation of target cells from other types of somatic cells. The most important and interesting advantage of this technique is that direct lineage conversion could bypass the pluripotent state, and therefore might reduce the risk of tumor formation. In addition, the process of direct lineage conversion requires less time than does the conventional differentiation by iPSCs or ESCs.

Currently, using defined transcription factors, direct lineage conversion has been applied to generate various cell types, including neurons,Citation73Citation75 kidney cells,Citation76 endocrine beta cells,Citation77 hepatocytes,Citation78 oligodendroglial cells,Citation79 as well as RPE cells.Citation23 Zhang et alCitation23 reported that defined transcription factors (cMyc, Mitf, Otx2, Rax, and Crx) could reprogram human fibroblasts into RPE cells by supplementation with retinoic acid and sonic hedgehog in a matrigel-based culture condition. These cells exhibit specific morphological and molecular features of RPE lineage and are capable of pigmentation. The most significant weakness in this study was that the suspected cells were not further evaluated by a functional test. However, this study still provided a novel direction to learn the nature of cellular identity and plasticity of RPE lineage, and also conducted a new approach to obtain functional RPE cells for regenerative medicine.

MSC-based microenvironmental regulation

Oxidative stress, overexpression of inflammatory cytokines, and retinal nutritional deficiency are some common mechanisms of AMD.Citation5 MSCs have various biological effects,Citation28 including secreting neurotrophins, promoting angiogenesis, regulating immune responses, inhibiting apoptosis, promoting extracellular matrix remodeling, and activating adjacent host stem cells. Furthermore, due to the low immunogenicity, MSCs are also ideal vehicles for introducing exogenous neurotrophic genes which could be expressed in the host retina. Therefore, MSCs are excellent candidates for dry AMD treatment.

On the basis of different origins, MSCs can be classified into bone marrow-derived MSCs (BM-MSCs), umbilical cord blood MSCs, placenta-derived MSCs, adipose-derived MSCs, and so on. BM-MSCs are the most well-studied groups of MSCs. This section will focus on the recent applications of BM-MSCs in AMD therapy.

Roles of BM-MSCs on retinal microenvironmental regulation

BM-MSCs can secrete neurotrophins

Inoue et alCitation80 reported that conditioned medium of BM-MSCs could inhibit photoreceptor apoptosis in vitro. After intravitreal injection of BM-MSCs, photoreceptor apoptosis was also delayed, and retinal function was slightly restored in RCS rats. These results indicated that soluble factors secreted by BM-MSCs may inhibit photoreceptor apoptosis. In another study, Zhang and WangCitation81 found that intravitreally injected BM-MSCs could express brain-derived neurotrophic factor (BDNF) and protect the outer nuclear layer in light-damaged retina. Xu et alCitation29,Citation30 also reported that MSCs could secret basic fibroblast growth factor and exhibit neuroprotective effects in light-damaged retina. Importantly, not only intravitreal injection but also intravenous injection of MSC could achieve retinal protective effects. Wang et alCitation82 reported that intravenous injection of 1×106 MSCs increased the survival of photoreceptors and restored the visual functions in RCS rats. Reverse transcriptase polymerase chain reaction and immunohistochemistry suggested that the protective effects were attributed to the retinal neurotrophins secreted by MSCs.

BM-MSCs can alleviate retinal inflammation

Xu et alCitation29,Citation30 found that intravitreal injection of BM-MSCs could suppress microglia activation, thereby reducing the retinal injury.

BM-MSCs can inhibit neuronal apoptosis

Otani et alCitation83 showed that retinal antiapoptotic genes were significantly upregulated after intravitreal injection of BM-MSCs. These genes included low-molecular-weight heat shock proteins and transcription factors.

BM-MSCs integrate into the host retina

Arnhold et alCitation84 found that intravitreal injection of BM-MSCs could significantly protect photoreceptors in rhodopsin knockout retinitis pigmentosa mice. They also showed that the transplanted BM-MSCs were well integrated into the RPE layer and the neurosensory layer of the host retina.

Notably, 1) the survival or integration of MSCs originated from different tissues might be very diverse. Intravitreally injected UCB-MSCs rarely migrated to the retina and only survived for 3 weeks,Citation85 whereas BM-MSCs survived for up to 20 weeks and had a good integration ability.Citation86 2) The neuroprotective effects of MSCs might be different between species. A study conducted by Levkovitch-Verbin et alCitation87 revealed that protection of retinal ganglion cells was merely noticed in human BM-MSCs, but not in rat BM-MSCs. 3) Methods for transplantation always relate to the experimental outcomes. Tzameret et alCitation86 compared the effects of intravitreal injection and subretinal injection in RCS rats. They found that the therapeutic effects lasted 12 and 20 weeks, respectively. The b-wave amplitudes in the electroretinogram were 56.4 μV in the intravitreal injection group and 66.2 μV in the subretinal injection group. 4) Retinal microenvironments in the host eyes also affect the functions of MSCs.

On the basis of the successful works in vivo, several Phase I/II clinical trials of MSCs were prudently conducted by some leading ophthalmologists. In 2005, Kumar et alCitation88 reported the outcomes of intravitreal injections of autologous BM-MSCs in 25 patients with dry AMD and retinitis pigmentosa. The mild improvement in BCVA was noticed after 1 or 3 months of injection. In 2010, Jonas et alCitation89 (registration number NCT1068561) reported the primary outcomes of three cases that received BM-MSC intravitreal injection (including one case of dry AMD). The initial BCVA of patients was poor in terms of light perception (poor light positioning). Twelve months after BM-MSC injection, no significant improvement in visual acuity and no serious complications were observed. The only effect was fluctuations of intraocular pressure (15–30 mmHg) at 4 weeks after treatment. Siqueira et alCitation90 intravitreally injected 1×107 BM-MSCs per eye in three retinitis pigmentosa patients and two cone–rod dystrophy patients. The results indicated that the visual acuities improved more than one row in four patients after 1 week and that these improvements were maintained at the end of the follow-up. Electrophysiological recordings of two patients were mildly improved. However, no significant changes in angiography, optical coherence tomography, and visual field were observed. Although the current clinical trials have not shown promising results, we must bear in mind the following: 1) Patients enrolled were relatively old, and their BM-MSCs have limited proliferative capacity and viability and 2) the patients were in advanced stages of disease. Therefore, vision recovery in these patients is sometimes difficult.

Effects of gene-modified MSCs

As an alternative to a viral vector, the application of stem cells to transfer specific genes is under investigation in various organs, including the eye.Citation31 Guan et alCitation32 found that after transplanting gene-modified MSCs into the subretinal spaces of sodium iodate-damaged eyes, a significant increase in erythropoietin was noticed and gene-modified MSCs showed stronger protective effects on retinal neurons than did conventional MSCs. Machalinska et alCitation33 also found that gene-modified MSCs stably expressing the NT-4 gene could migrate to the retinal damage area and protect the damaged cells. More importantly, gene-modified MSCs could upregulate the signals and transcription factors related to cell survival, such as crystallin β–γ superfamily members. In addition, gene-modified MSCs also increased the expression of proteins related to visual perception, visual signal reception, and eye development. In another study, Park et alCitation34 evaluated the integration ability of gene-modified BM-MSCs and their BDNF secretion in vivo. They found that approximately 15.7% of the MSCs integrated into the retina after 4 weeks. The protein and mRNA levels of BDNF were greatly increased in the host retina. The function of gene-modified MSCs is largely dependent on the genes they deliver. Choosing suitable genes and delivery protocols will enable us to establish a new direction for ADM treatment.

Prospects

In-depth studies on the biological characteristics of stem cell-derived RPEs, differentiation protocols, and transplantation methods are gradually changing the current stem cell-based therapy from a dream to reality. However, there are still several obstacles before their clinical application. Transplanted RPE cells showed limited adhesion and survival in human eyes, and aged Bruch’s membrane did not likely support adhesion, survival, differentiation, and function of grafted RPE cells.Citation91Citation94 Therefore, the use of genetic engineering to overexpress integrins or integrin activators in the RPE cellsCitation95Citation97 or the use of RPE cells growing on scaffolds might show promising prospects. Second, although subretinal space was once considered to have immune privilege, studies also have indicated that the long-term survival of the transplanted cells in the host eyes still required immune suppression.Citation25Citation27 Thus, the course of immunosuppression and the drugs used for immunosuppression have to be further discussed.

Acknowledgments

This study was supported by the International Cooperation Project of Henan Province (2013GH11), the National Natural Science Foundation of China (No 81371017), and the Key Project of Science Research of Henan Province Education Committee (No 13A320427).

Disclosure

The authors report no conflicts of interest in this work.

References

  • FriedmanDSO’ColmainBJMuñozBEye Diseases Prevalence Research GroupPrevalence of age-related macular degeneration in the United StatesArch Ophthalmol2004122456457215078675
  • VingerlingJRDielemansIHofmanAThe prevalence of age-related maculopathy in the Rotterdam StudyOphthalmology199510222052107862408
  • KleinRKnudtsonMDLeeKEAge-period-cohort effect on the incidence of age-related macular degeneration: the Beaver Dam Eye StudyOphthalmology200811591460146718762073
  • KleinRKleinBELeeKEChanges in visual acuity in a population over a 15-year period: the Beaver Dam Eye StudyAm J Ophthalmol2006142453954917011842
  • ParmeggianiFRomanoMRCostagliolaCMechanism of inflammation in age-related macular degenerationMediators Inflamm2012201254678623209345
  • Age-Related Eye Disease Study Research GroupA randomized, placebo-controlled, clinical trial of high-dose supplementation with vitamins C and E, beta carotene, and zinc for age-related macular degeneration and vision loss: AREDS report no. 8Arch Ophthalmol2001119101417143611594942
  • KawasakiHSuemoriHMizusekiKGeneration of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activityProc Natl Acad Sci U S A20029931580158511818560
  • HarutaMSasaiYKawasakiHIn vitro and in vivo characterization of pigment epithelial cells differentiated from primate embryonic stem cellsInvest Ophthalmol Vis Sci20044531020102514985325
  • LundRDWangSKlimanskayaIHuman embryonic stem cell-derived cells rescue visual function in dystrophic RCS ratsCloning Stem Cells2006818919917009895
  • KlimanskayaIHippJRezaiKADerivation and comparative assessment of retinal pigment epithelium from human embryonic stem cells using transcriptomicsCloning Stem Cells2004621724515671670
  • VaajasaariHIlmarinenTJuuti-UusitaloKToward the defined and xeno-free differentiation of functional human pluripotent stem cell-derived retinal pigment epithelial cellsMol Vis20111755857521364903
  • ZhangYSLuZYYuYDerivation, culture and retinal pigment epithelial differentiation of human embryonic stem cells using human fibroblast feeder cellsJ Assist Reprod Genet201229873574422661130
  • ParkUCChoMSParkJHSubretinal transplantation of putative retinal pigment epithelial cells derived from human embryonic stem cells in rat retinal degeneration modelClin Exp Reprod Med201138421622122384445
  • MeyerJSShearerRLCapowskiEEModeling early retinal development with human embryonic and induced pluripotent stem cellsProc Natl Acad Sci U S A200910639166981670319706890
  • CarrAJVuglerAAHikitaSTProtective effects of human iPS-derived retinal pigment epithelium cell transplantation in the retinal dystrophic ratPLoS One2009412e815219997644
  • BuchholzDEHikitaSTRowlandTJDerivation of functional retinal pigmented epithelium from induced pluripotent stem cellsStem Cells200927102427243419658190
  • HuangCZhangJAoMCombination of retinal pigment epithelium cell-conditioned medium and photoreceptor outer segments stimulate mesenchymal stem cell differentiation toward a functional retinal pigment epithelium cell phenotypeJ Cell Biochem2012113259059821948619
  • IkedaHOsakadaFWatanabeKGeneration of Rx+/Pax6+ neural retinal precursors from embryonic stem cellsProc Natl Acad Sci U S A200510232113311133616076961
  • OsakadaFIkedaHSasaiYTakahashiMStepwise differentiation of pluripotent stem cells into retinal cellsNat Protoc20094681182419444239
  • KamaoHMandaiMOkamotoSCharacterization of human induced pluripotent stem cell-derived retinal pigment epithelium cell sheets aiming for clinical applicationStem Cell Reports20142220521824527394
  • BrandlCZimmermannSJMilenkovicVMIn-depth characterization of retinal pigment epithelium (RPE) cells derived from human induced pluripotent stem cells (hiPSC)Neuromolecular Med201416355156424801942
  • KokkinakiMSahibzadaNGolestanehNHuman 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 RPEStem Cells201129582583521480547
  • ZhangKLiuGHYiFDirect conversion of human fibroblasts into retinal pigment epithelium-like cells by defined factorsProtein Cell201451485824474194
  • SchwartzSDHubschmanJPHeilwellGEmbryonic stem cell trials for macular degeneration: a preliminary reportLancet2012379981771372022281388
  • Del PrioreLVIshidaOJohnsonEWTriple immune suppression increases short-term survival of porcine fetal retinal pigment epithelium xenograftsInvest Ophthalmol Vis Sci20034494044405312939327
  • LaiCCGourasPDioKLocal immunosuppression prolongs survival of RPE xenografts labeled by retroviral gene transferInvest Ophthalmol Vis Sci200041103134314110967075
  • HeSWangHMOgdenTERyanSJTransplantation of cultured human retinal pigment epithelium into rabbit subretinaGraefes Arch Clin Exp Ophthalmol1993231127377428299983
  • SiqueiraRCVoltarelliJCMessiasAMPossible mechanisms of retinal function recovery with the use of cell therapy with bone marrow-derived stem cellsArq Bras Oftalmol201073547447921225138
  • XuWWangXXuGBasic fibroblast growth factor expression is implicated in mesenchymal stem cells response to light-induced retinal injuryCell Mol Neurobiol20133381171117924030359
  • XuWWangXXuGLight-induced retinal injury enhanced neurotrophins secretion and neurotrophic effect of mesenchymal stem cells in vitroArq Bras Oftalmol201376210511023828471
  • HuangLXuWXuGTransplantation of CX3CL1-expressing mesenchymal stem cells provides neuroprotective and immunomodulatory effects in a rat model of retinal degenerationOcul Immunol Inflamm201321427628523718544
  • GuanYCuiLQuZSubretinal transplantation of rat MSCs and erythropoietin gene modified rat MSCs for protecting and rescuing degenerative retina in ratsCurr Mol Med20131391419143123971737
  • MachalinskaAKawaMPPius-SadowskaELong-term neuroprotective effects of NT-4-engineered mesenchymal stem cells injected intravitreally in a mouse model of acute retinal injuryInvest Ophthalmol Vis Sci201354138292830524265016
  • ParkHYKimJHSun KimHStem cell-based delivery of brain-derived neurotrophic factor gene in the rat retinaBrain Res20121469102322750585
  • MachemerRSteinhorstUHRetinal separation, retinotomy, and macular relocation: II. A surgical approach for age-related macular degeneration?Graefes Arch Clin Exp Ophthalmol1993231116356418258397
  • de JuanEJrLoewensteinABresslerNMAlexanderJTranslocation of the retina for management of subfoveal choroidal neovascularization II: a preliminary report in humansAm J Ophthalmol199812556356469625547
  • LaiJCLapoliceDJStinnettSSVisual outcomes following macular translocation with 360-degree peripheral retinectomyArch Ophthalmol2002120101317132412365910
  • TakeuchiKKachiSIwataEIshikawaKTerasakiHVisual function 5 years or more after macular translocation surgery for myopic choroidal neovascularisation and age-related macular degenerationEye (Lond)2012261516022173070
  • ChenFKPatelPJUppalGSTufailACoffeyPJDa CruzLLong-term outcomes following full macular translocation surgery in neovascular age-related macular degenerationBr J Ophthalmol201094101337174320494910
  • GeliskenFVoelkerMSchwabeRFull macular translocation versus photodynamic therapy with verteporfin in the treatment of neovascular age-related macular degeneration: 1-year results of a prospective, controlled, randomized pilot trial (FMT-PDT)Graefes Arch Clin Exp Ophthalmol200724581085109517219106
  • Falkner-RadlerCIKrebsIGlittenbergCHuman retinal pigment epithelium (RPE) transplantation: outcome after autologous RPE-choroid sheet and RPE cell-suspension in a randomized clinical studyBr J Ophthalmol201195337037520610478
  • MacLarenREUppalGSBalagganKSAutologous transplantation of the retinal pigment epithelium and choroid in the treatment of neovascular age-related macular degenerationOphthalmology2007114356157017324698
  • JoussenAMHeussenFMJoeresSAutologous translocation of the choroid and retinal pigment epithelium in age-related macular degenerationAm J Ophthalmol20061421173016815247
  • JoussenAMHow complete is successful? “Autologous retinal pigment epithelium and choriod translocation in patients with exsudative age-related macular degeneration: a short-term follow-up” by Jan van Meurs and P.R. van BiesenGraefes Arch Clin Exp Ophthalmol20032411296696714624301
  • BinderSKrebsIHilgersRDOutcome of transplantation of autologous retinal pigment epithelium in age-related macular degeneration: a prospective trialInvest Ophthalmol Vis Sci200445114151416015505069
  • VuglerACarrAJLawrenceJElucidating the phenomenon of HESC-derived RPE: anatomy of cell genesis, expansion and retinal transplantationExp Neurol2008214234736118926821
  • TezelTHDel PrioreLVKaplanHJReengineering of aged Bruch’s membrane to enhance retinal pigment epithelium repopulationInvest Ophthalmol Vis Sci20044593337334815326159
  • MartinMJMuotriAGageFVarkiAHuman embryonic stem cells express an immunogenic nonhuman sialic acidNat Med200511222823215685172
  • SakamotoNTsujiKMuulLMBovine apolipoprotein B-100 is a dominant immunogen in therapeutic cell populations cultured in fetal calf serum in mice and humansBlood2007110250150817395779
  • LuBMalcuitCWangSLong-term safety and function of RPE from human embryonic stem cells in preclinical models of macular degenerationStem Cells2009272126213519521979
  • EirakuMTakataNIshibashiHSelf-organizing optic-cup morphogenesis in three-dimensional cultureNature20114727341515621475194
  • ZhuYCaridoMMeinhardtAThree-dimensional neuroepithelial culture from human embryonic stem cells and its use for quantitative conversion to retinal pigment epitheliumPLoS One201381e5455223358448
  • ChoMYKimSJKuSYGeneration of retinal pigment epithelial cells from human embryonic stem cell-derived spherical neural massesStem Cell Res20129210110922683799
  • LambaDAKarlMOWareCBEfficient generation of retinal progenitor cells from human embryonic stem cellsProc Natl Acad Sci U S A200610334127691277416908856
  • IdelsonMAlperRObolenskyADirected differentiation of human embryonic stem cells into functional retinal pigment epithelium cellsCell Stem Cell20095439640819796620
  • SakamiSEtterPRehTAActivin signaling limits the competence for retinal regeneration from the pigmented epitheliumMech Dev20081251–210611618042353
  • BuchholzDEPenningtonBOCrozeRHHinmanCRCoffeyPJCleggDORapid and efficient directed differentiation of human pluripotent stem cells into retinal pigmented epitheliumStem Cells Transl Med20132538439323599499
  • OsakadaFJinZBHiramiYIn vitro differentiation of retinal cells from human pluripotent stem cells by small-molecule inductionJ Cell Sci2009122173169317919671662
  • TakahashiKYamanakaSInduction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factorsCell2006126466367616904174
  • YuJVodyanikMASmuga-OttoKInduced pluripotent stem cell lines derived from human somatic cellsScience200731858581917192018029452
  • HiramiYOsakadaFTakahashiKGeneration of retinal cells from mouse and human induced pluripotent stem cellsNeurosci Lett2009458312613119379795
  • WestenskowPDMorenoSKKrohneTUUsing flow cytometry to compare the dynamics of photoreceptor outer segment phagocytosis in iPS-derived RPE cellsInvest Ophthalmol Vis Sci201253106282629022871841
  • KrohneTUWestenskowPDKuriharaTGeneration of retinal pigment epithelial cells from small molecules and OCT4 reprogrammed human induced pluripotent stem cellsStem Cells Transl Med2012129610922532929
  • HuQFriedrichAMJohnsonLVCleggDOMemory in induced pluripotent stem cells: reprogrammed human retinal-pigmented epithelial cells show tendency for spontaneous redifferentiationStem Cells201028111981199120882530
  • RayaARodríguez-PizàIGuenecheaGDisease-corrected haematopoietic progenitors from Fanconi anemia induced pluripotent stem cellsNature20094607251535919483674
  • GongLWuQSongBDifferentiation of rat mesenchymal stem cells transplanted into the subretinal space of sodium iodate-injected ratsClin Experiment Ophthalmol200836766667118983552
  • TomitaMAdachiYYamadaHBone marrow-derived stem cells can differentiate into retinal cells in injured rat retinaStem Cells200220427928312110696
  • TropepeVColesBLChiassonBJRetinal stem cells in the adult mammalian eyeScience200028754602032203610720333
  • ArutaCGiordanoFDe MarzoAIn vitro differentiation of retinal pigment epithelium from adult retinal stem cellsPigment Cell Melanoma Res201124123324021232026
  • CiceroSAJohnsonDReyntjensSCells previously identified as retinal stem cells are pigmented ciliary epithelial cellsProc Natl Acad Sci U S A2009106166685669019346468
  • BernardosRLBarthelLKMeyersJRLate-stage neuronal progenitors in the retina are radial Müller glia that function as retinal stem cellsJ Neurosci200727267028704017596452
  • SongWTZhangXYXiaXBAtoh7 promotes the differentiation of retinal stem cells derived from Müller cells into retinal ganglion cells by inhibiting Notch signalingStem Cell Res Ther2013449423945288
  • ChandaSAngCEDavilaJGeneration of induced neuronal cells by the single reprogramming factor ASCL1Stem Cell Reports20143228229625254342
  • KarowMSchichorCBeckervordersandforthRBerningerBLineage-reprogramming of pericyte-derived cells of the adult human brain into induced neuronsJ Vis Exp201487
  • WapinskiOLVierbuchenTQuKHierarchical mechanisms for direct reprogramming of fibroblasts to neuronsCell2013155362163524243019
  • TakasatoMVanslambrouckJMLittleMHReprogramming somatic cells to a kidney fateSemin Nephrol201434446248025217274
  • Cavelti-WederCLiWWeirGCZhouQDirect lineage conversion of pancreatic exocrine to endocrine beta cells in vivo with defined factorsMethods Mol Biol2014115024726224744004
  • HuangPZhangLGaoYDirect reprogramming of human fibroblasts to functional and expandable hepatocytesCell Stem Cell201414337038424582927
  • YangNZucheroJBAhleniusHGeneration of oligodendroglial cells by direct lineage conversionNat Biotechnol201331543443923584610
  • InoueYIriyamaAUenoSSubretinal transplantation of bone marrow mesenchymal stem cells delays retinal degeneration in the RCS rat model of retinal degenerationExp Eye Res200785223424117570362
  • ZhangYWangWEffects of bone marrow mesenchymal stem cell transplantation on light-damaged retinaInvest Ophthalmol Vis Sci20105173742374820207980
  • WangSLuBGirmanSNoninvasive stem cell therapy in a rat model for retinal degeneration and vascular pathologyPLoS One201052e920020169166
  • OtaniADorrellMIKinderKRescue of retinal degeneration by intravitreally injected adult bone marrow-derived lineage-negative hematopoietic stem cellsJ Clin Invest2004114676577415372100
  • ArnholdSAbsengerYKleinHTransplantation of bone marrow-derived mesenchymal stem cells rescue photoreceptor cells in the dystrophic retina of the rhodopsin knockout mouseGraefes Arch Clin Exp Ophthalmol2007245314422
  • HillAJZwartITamHHHuman umbilical cord blood-derived mesenchymal stem cells do not differentiate into neural cell types or integrate into the retina after intravitreal grafting in neonatal ratsStem Cells Dev200918339940918665766
  • TzameretASherIBelkinMTransplantation of human bone marrow mesenchymal stem cells as a thin subretinal layer ameliorates retinal degeneration in a rat model of retinal dystrophyExp Eye Res201411813514424239509
  • Levkovitch-VerbinHSadanOVanderSIntravitreal injections of neurotrophic factors secreting mesenchymal stem cells are neuroprotective in rat eyes following optic nerve transectionInvest Ophthalmol Vis Sci201051126394640020926814
  • KumarAPahwaVKTandonRKumarLMohantySUse of autologous bone marrow derived stem cells for rehabilitation of patients with dry age related macular degeneration and retinitis pigmentosa: Phase-1 clinical trialIndian J Med Paediatr Oncol200526Suppl 31214
  • JonasJBWitzens-HarigMArsenievLIntravitreal autologous bone-marrow-derived mononuclear cell transplantationActa Ophthalmol2010884e131e13219604156
  • SiqueiraRCMessiasAVoltarelliJCIntravitreal injection of autologous bone marrow-derived mononuclear cells for hereditary retinal dystrophy: a Phase I trialRetina20113161207121421293313
  • TezelTHKaplanHJDel PrioreLVFate of human retinal pigment epithelial cells seeded onto layers of human Bruch’s membraneInvest Ophthalmol Vis Sci1999404674769950607
  • Del PrioreLVTezelTHReattachment rate of human retinal pigment epithelium to layers of human Bruch’s membraneArch Ophthalmol19981163353419514487
  • GullapalliVKSuginoIKVan PattenYRetinal pigment epithelium resurfacing of aged submacular human Bruch’s membraneTrans Am Ophthalmol Soc2004102123137 discussion 137–13815747751
  • SunKCaiHTezelTHBruch’s membrane aging decreases phagocytosis of outer segments by retinal pigment epitheliumMol Vis2007132310231918199972
  • AfshariFTKwokJCAndrewsMRIntegrin activation or alpha 9 expression allows retinal pigmented epithelial cell adhesion on Bruch’s membrane in wet age-related macular degenerationBrain201013344846420159768
  • AfshariFTFawcettJWImproving RPE adhesion to Bruch’s membraneEye200923101890189319151642
  • FangIMYangCHYangCMChenMSOverexpression of integrin alpha6 and beta4 enhances adhesion and proliferation of human retinal pigment epithelial cells on layers of porcine Bruch’s membraneExp Eye Res200988122118955047