Next Article in Journal
Age Related Osteoporosis: Targeting Cellular Senescence
Next Article in Special Issue
Pleiotropic Roles of Scavenger Receptors in Circadian Retinal Phagocytosis: A New Function for Lysosomal SR-B2/LIMP-2 at the RPE Cell Surface
Previous Article in Journal
Pediococcus acidilactici CECT9879 (pA1c) Counteracts the Effect of a High-Glucose Exposure in C. elegans by Affecting the Insulin Signaling Pathway (IIS)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circadian Regulation of Retinal Pigment Epithelium Function

Department of Pharmacology & Toxicology and Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA 30310-1495, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(5), 2699; https://doi.org/10.3390/ijms23052699
Submission received: 26 January 2022 / Revised: 16 February 2022 / Accepted: 21 February 2022 / Published: 28 February 2022

Abstract

:
The retinal pigment epithelium (RPE) is a single layer of cells located between the choriocapillaris vessels and the light-sensitive photoreceptors in the outer retina. The RPE performs physiological processes necessary for the maintenance and support of photoreceptors and visual function. Among the many functions performed by the RPE, the timing of the peak in phagocytic activity by the RPE of the photoreceptor outer segments that occurs 1–2 h. after the onset of light has captured the interest of many investigators and has thus been intensively studied. Several studies have shown that this burst in phagocytic activity by the RPE is under circadian control and is present in nocturnal and diurnal species and rod and cone photoreceptors. Previous investigations have demonstrated that a functional circadian clock exists within multiple retinal cell types and RPE cells. However, the anatomical location of the circadian controlling this activity is not clear. Experimental evidence indicates that the circadian clock, melatonin, dopamine, and integrin signaling play a key role in controlling this rhythm. A series of very recent studies report that the circadian clock in the RPE controls the daily peak in phagocytic activity. However, the loss of the burst in phagocytic activity after light onset does not result in photoreceptor or RPE deterioration during aging. In the current review, we summarized the current knowledge on the mechanism controlling this phenomenon and the physiological role of this peak.

1. Introduction

Circadian rhythms are important features of living organisms. These rhythms are endogenously generated by molecular clocks (i.e., the circadian clocks) that have a period close to 24 h. This circadian oscillation is generated by a transcriptional translational feedback loop that involves several “clock” genes and their products (Figure 1). Numerous studies have also shown that circadian clocks are present in several ocular tissues (e.g., retina, cornea, retinal pigment epithelium, etc.) where they control critical physiological functions (see [1] for a recent review and references within). Additional studies have also reported that dysfunction of the circadian clock and its outputs (i.e., melatonin, dopamine, etc.) in the eye may adversely affect these tissues [2,3,4,5,6,7,8,9,10].
The retinal pigment epithelium (RPE) is a single layer of cells located between the choriocapillaris vessels and the light-sensitive photoreceptors in the outer retina. The RPE performs physiological processes necessary for the maintenance and support of photoreceptors and visual function. Among the several functions of the RPE, the role in the continuous renewal of the light-sensitive outer segment of photoreceptors is critical for the photoreceptor health [11]. Photoreceptors synthesize new outer segment components and form new outer segment disks. A process commonly termed disk shedding compensates for this addition during which RPE cells, in collaboration with photoreceptors, remove the most distal tip of rod and cone outer segments (POS) [12,13]. The POS sheds and then is promptly engulfed by the RPE and degraded with this tissue. Previous studies show that the lack of phagocytosis by the RPE leads to the accumulation of POS in the subretinal space and consequently to photoreceptor degeneration [14]. The present review aimed to summarize the current knowledge about the circadian regulation of RPE function and how disruption of the circadian function may lead to many pathologies.

2. Regulation of RPE Function In Vivo

The process of photoreceptor outer segment shedding and phagocytosis follows a diurnal rhythm characterized by a burst/peak in the level of phagocytosis 1–2 h after the onset of light [15]. This rhythm persists under constant darkness [16,17] or light [18], after the optic nerve has been severed [19], and once the master circadian clock located in the brain has been ablated [20]. Because of this experimental evidence, it was proposed that the rhythms in photoreceptor disc shedding, and phagocytosis were under the control of circadian clocks located within the eye [16,19,20]. Indeed, later studies demonstrated the presence of circadian clocks in the retina [21], within the photoreceptors [22,23], the inner retina [23,24], and within the RPE [25,26]. However, it is still unclear whether the rhythm in phagocytosis by the RPE is controlled by the circadian clock located in the retina, in the RPE, or both. It is also worth noting that although most of the studies on RPE phagocytic activity have focused on the process controlling phagocytosis in the rod photoreceptors, a circadian process has also been described for the cone photoreceptors [27,28]. Since the burst in phagocytic activity is present in many vertebrate species (including humans) and other experimental conditions, it was hypothesized that the presence and timing of the peak in the phagocytic activity must play an important role in the health of the photoreceptors and the RPE [1]. However, it is important to mention that although several studies have indicated that phagocytic activity by the RPE play and important role in maintain a healthy retina in humans (Table 1) no study so far has indicated that dysfunction the phagocytic rhythm in humans is a cofactor in the development of human retinal pathologies.
Hence, in the last twenty years, many laboratories investigated the molecular mechanisms controlling the burst in the phagocytic activity and then have produced a new animal model to prove that the presence of this peak was indeed important for the overall health of the RPE and the photoreceptors. These studies reported a lack the activation of key agents of phagocytic signaling, including focal adhesion kinase (FAK) [29,30,31], MerTK [29,30], and Rac1 GTPase, a potent F-actin regulator [31,32], to be the underlying cause of the disturbed phagocytosis process by the RPE. In 2012, Ruggiero et al. [33] also reported that the rods expose a conserved phosphatidylserine domain at their distal tip, which serves as an “eat me” signal at the light onset. Interestingly, this phosphatidylserine exposure is not rhythmic in mice lacking the diurnal rhythm of RPE phagocytosis due to a lack of αvβ5 integrin receptor or its ligand Milk Fat Globule-EGF factor 8 protein (MFG-E8), thus suggesting that the key signal initiating the burst of phagocytic activity is from the RPE. Other studies have also implicated dopamine and melatonin signaling in this process since Dopamine 2 Receptors (D2R) knock-out (KO) mice do not show a daily peak in phagocytic activity [31], and melatonin receptor KO mice show a disruption in the timing of the peak [34]. However, it is important to mention that, in all these studies, the total daily phagocytic activity was not different from the wild-type mice. Other investigations have also revealed that mice lacking myosin VIIa, Annexin A2, and lysosomal protein melanoregulin showed a normal phagocytic peak but delay in trafficking from the apical to the basal region of the RPE [35,36].
To gather further insights into the molecular mechanism and signaling pathways controlling the circadian rhythm of the phagocytic activity in the RPE, several studies have conducted transcriptomic profiling of this tissue at various time points before, during, and after the phagocytosis process. In one such study in 2013, Mustafi et al. [37] found that several genes (Dgki, Itpr1, Pik3r1, Lamp2 and Pla2g2) involved in polyphosphoinositide signaling are upregulated in mice RPE 1.5 h. after the onset of light. Along with that, the promoter region of these genes also contains CLOCK: BMAL1 and ROR binding motifs, thus reinforcing the notion that the circadian clock in the RPE is modulating the phosphoinositide signaling. An additional study using RNA-sequencing and pathway analysis reported that approximately 20% of the RPE transcriptome is under circadian regulation [38]. This study also showed that one hour after the subjective light onset, pathways associated with RPE phagocytosis, including integrin signaling, cAMP signaling, focal adhesion, epithelial adherence junction signaling, mitochondrial phosphorylation, and protein phosphorylation, are upregulated in the RPE [38]. The circadian regulation of the RPE transcriptome also involves the regulation of metabolic pathways since the transcription of several genes involved in ATP (Adenosine triphosphate) production, fat metabolism, and other metabolic pathways are under circadian control [39]. In the middle of the night, transcripts involved in the mitochondrial ETC (electron transport chain), TCA (tricarboxylic acid) cycle, glycolysis, and glycogen metabolism were higher, suggesting a greater need for energy production at this time point in the RPE. Additionally, an up-regulation of genes implicated in the glycerophospholipid metabolism pathway was also observed at this time. Glycerophospholipids are the building blocks of membrane lipids. An upregulation in the metabolism of glycerophospholipids at night might suggest that they could be transported to photoreceptor cells to form new outer space segments [40] as soon as the exposed outer segments are shed. Conversely, the fatty acid degradation pathway was also upregulated during the day as the RPE digests the lipid layer of the ingested POS.
Although several studies have investigated the effects of clock genes removal in the retina, only two investigations have examined the effect of clock gene removal on the circadian regulation of phagocytic activity. In the first study, Per1/Per2 global KO blunted the peak in phagocytic activity, and 57 genes involved in photoreceptor phagocytosis were downregulated in the RPE [41]. In the second study, DeVera et al. [42] developed an RPE-specific Bmal1 KO mouse model. They demonstrated that the circadian clock in the RPE controls the daily diurnal peak in phagocytosis of POS since, in retina specific Bmal1 KO, the daily rhythm in phagocytic activity was not affected by the removal of this gene. In contrast, the removal of Bmal1 from the RPE abolished the daily rhythm [42].
Table 1. List of protein that have been associated with the daily rhythm in phagocytic activity and their involvement in human retinal diseases.
Table 1. List of protein that have been associated with the daily rhythm in phagocytic activity and their involvement in human retinal diseases.
Function in RPEAnimal ModelHuman Retinal Disease
MerTK receptorOuter segment binding & internalizationRCS rat [43,44]
Merkd mouse [45]
Retinitis pigmentosa, rod-cone dystrophy [46,47,48,49,50]
Gas6, Protein SMerTK ligandsGas6 double KO and ProS1 [51] Diabetic Retinopathy and macular edema [52]
ανβ5 integrin receptorOuter segment binding,
Control the diurnal rhythm in peak of phagocytosis
β5−/− mouse [29]unknown
MFG-E8ανβ5 integrin ligand
Control the diurnal rhythm in peak of phagocytosis
MFG-E8−/− mouse [30]unknown
Dopamine receptor 2Controls the rhythm in RPE circadian clocks, light adaptation, peak of phagocytosis after light onsetD2R KO mouse [31]unkown
Melatonin receptor 1 and 2Control the timing of the peak of phagocytosisMT1 & MT2 KO mouse [34]
RPE specific Bmal1 KOControl the diurnal rhythm in peak of phagocytosisRPEcre; Bmal1fl/fl [42]unknown
Per1/Per2 global KOControls the amplitude of the peak of phagocytosisPer1−/−Per2Brdm1 [41]unknown

3. Regulation of RPE Function In Vitro

In the last few years, a few methods have been introduced to culture primary RPE cells from many species, including humans [53,54]. Some procedures include an RPE sheet peeled from the choroidal cup for large animals [55,56]. However, isolating RPE sheets from small animals, such as the mouse, is still a challenge [25,57]. As an alternative to primary RPE culture, a few immortalized RPE cell lines have been produced. The most used RPE cell line is ARPE-19, which was established from cells isolated from the enucleated globes of a 19-year-old male donor [58]. In addition, the RPE-J cell line is derived from a 7-day-old Long-Evans rat and is commonly used for non-human RPE studies [59]. While these cell lines show similar phenotypical features as native RPE cells, the cell lines have some physiological and morphological differences from native cells, such as a lack of pigmentation and turnover [60,61,62].
The first circadian clock oscillation in in vitro RPE cells was observed in human RPE cell lines (hRPE). In this study, hRPE cells were first synchronized with forskolin, and then the cells were collected every 6 h. The authors reported Per1 and Per2 mRNA levels were rhythmically transcribed for at least three circadian cycles [63]. Then, Yoshikawa et al. [64], using a Bmal1-luciferase bioluminescence reporter system, demonstrated a circadian rhythm in Bmal1-luciferase bioluminescence for 7 days in hRPE cells. More recent findings also revealed that several genes involved in RPE phagocytic activity are also expressed in a circadian manner in the ARPE19 cell line [65]. This research team also reported that the culture methodology (dispersed vs. monolayer) might affect clock gene rhythmic expression amplitude in ARPE19 cells.
The recent introduction of bioluminescence reporter technologies and the generation of transgenic mice in which bioluminescence can be recorded from tissue explants have led to significant advances in understanding the mammalian circadian system [66]. Using these newly generated transgenic mice, circadian rhythms have been observed in the retina, photoreceptor, cornea, iris-ciliary body, and RPE explants collected from PERIOD2::LUCIFERASE (PER2::LUC) mice eyes [23,24,25,26,67,68,69]. Using this mouse model, in 2010, Baba et al. showed that although a clear bioluminescence rhythm can be observed from isolated RPE cells, this rhythm has a larger amplitude and the RPE survive better when cultured together with choroid [25]. The phase and period of mice RPE PER2::LUC rhythm is slightly different from the retinal PER2::LUC rhythm (RPE vs. Retina Phases: ZT 16.5 h vs. ZT 12.4 h, light onset as ZT 0/dark onset as ZT 12; Periods: 23.9 h vs. 24.3 h). Moreover, this circadian rhythm can persist for more than fifty days with weekly medium exchanges [25]. Finally, it is worth mentioning that the age of the mice and the time of the day at which the tissue is explanted and prepared for the culture preparation may affect the circadian parameters, and thus it should be carefully considered [7,70].

4. Entrainment of the RPE Circadian Clock

A few studies have reported that light can entrain the circadian rhythms of the isolated retina and cornea from mice [24,71], but light does not appear to entrain the circadian rhythm of bioluminescence in the mouse RPE [25,72]. In the eye, melatonin and dopamine play an important role in the regulation and entrainment of circadian rhythms [1], and, as previously mentioned, melatonin signaling is involved in the regulation of the timing and amplitude of the morning phagocytic peak [31,33,73]. Several lines of evidence also suggest that dopamine and its receptors are involved in regulating rhythmic RPE function. For example, the inhibition of dopamine synthesis during the early part of the light phase induced a significant reduction of disk shedding and phagocytosis [74], and mice whose dopaminergic neurons have been destroyed by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) accumulate many residual bodies in the RPE [75].
This experimental evidence has led to the notion that these two neurohormones may be responsible for the entrainment of the RPE circadian clock. Indeed, Baba et al. [26] demonstrated that dopamine, but not melatonin, entrained the circadian rhythms in PER2::LUC bioluminescence in cultured RPE. The administration of exogenous dopamine (100 mM) entrains the circadian rhythm in the RPE in a phase-dependent manner. Moreover, it delays the RPE PER2::LUC rhythm during late night to early morning and phase advances PER2::LUC rhythm during the day [26]. Such an effect appears to be exclusively mediated by D2R signaling [26]. The mechanisms by which D2R affects entraining the circadian clock in the RPE are not yet understood since D2R signaling is negatively coupled to an adenylyl cyclase, which leads to reduced cAMP levels. Since previous studies have shown that increases in cAMP levels are indeed involved in the clock resetting mechanism by acting on cAMP responsive elements in the Period gene promoter [76,77,78] it must be concluded that it is unlikely that D2R receptor activation, which reduces cAMP levels, induces the phase shifts via the cAMP cascade. Moreover, it is worth mentioning that a previous study indicated that D2R signaling induces the phosphorylation of the cAMP Response Element Binding Protein (CREB) by activating Ca2+/calmodulin-dependent protein kinase [79] and the ERK1/2 signaling pathway [80,81,82]. ERK1/2 signaling has also been implicated in the mechanism of the entrainment of the master circadian clock [83,84,85,86]. Further studies also show that the 90RSK protein plays a key role in activating the ERK-CREB pathway [87,88]. Thus, we propose that the mechanism responsible for the entrainment of the RPE via D2R involves the ERK1/2/ pathway, as described in Figure 2.
An alternative mechanism for the entrainment of the circadian clock in the RPE was also proposed. According to Ikarashi et al. [72], cytosolic Ca2+ level shows circadian variation in RPE cells, and this circadian variation in Ca2+ levels is abolished in Bmal1 dominant-negative RPE cells. This study also showed that muscarinic receptor subtype, M3, is expressed in hRPE cells and the acetylcholine/carbachol-induced Ca2 elevation seems to be under circadian control. Finally, they reported that the administration of carbachol successfully phase-shifted Bmal1-luc rhythms in hRPE cells in a phase-dependent manner [72]. Additionally, histamine, another element known to increase cytosolic Ca2+ levels, phase-shifts Bmal1-luc rhythms in hRPE cells in a phase-dependent manner very similar to the carbachol-induced phase-shift [72]. This effect was abolished when histamine was treated with H1 receptor antagonists. In contrast, treatment with an H2 receptor agonist showed only a small effect. Thus, H1 receptor signaling is probably more dominant in mediating RPE circadian phase-shifts in hRPE cells [89]. Interestingly, another research group showed that three hours of a co-incubation with POS induced lysosomal marker, LAMP1, expression in cultured ARPE19 cells in a time-dependent manner [41]. Hence, this study indicates that the circadian clock regulates the phagocytic activity of the RPE, and photoreceptor outer binding availability and perhaps the phagocytic activity itself may provide feedback to the circadian clock in the RPE. Thus, although numerous studies have demonstrated that several factors are involved in the entrainment of the RPE circadian clock, further studies are needed to understand the mechanism controlling this phenomenon in the RPE fully.
Figure 2. Dopamine clock-resetting pathway in the RPE. Schematic illustration of the proposed mechanism by which D2R signaling phase shifts the circadian clock in the RPE. Activating these receptors by dopamine (DA) leads to MEK activation, then Erk1/2 (ERK), and finally the phosphorylation of p90RSK, which in turn phosphorylates CREB at Ser133, thereby activating the Per1/2 promoters. Alternatively, ERK can also induce Per1/2 transcription via the phosphorylation of CBP, causing it to bind to and activate the BMAL1:CLOCK [90].
Figure 2. Dopamine clock-resetting pathway in the RPE. Schematic illustration of the proposed mechanism by which D2R signaling phase shifts the circadian clock in the RPE. Activating these receptors by dopamine (DA) leads to MEK activation, then Erk1/2 (ERK), and finally the phosphorylation of p90RSK, which in turn phosphorylates CREB at Ser133, thereby activating the Per1/2 promoters. Alternatively, ERK can also induce Per1/2 transcription via the phosphorylation of CBP, causing it to bind to and activate the BMAL1:CLOCK [90].
Ijms 23 02699 g002

5. Is the Daily Burst in Phagocytic Activity Important for Photoreceptor and RPE Health?

As we have previously mentioned, several studies have supported the notion that the timing and presence of the daily peak in phagocytic activity play an important role in regulating the RPE and photoreceptor physiology and have speculated that a lack of this event may have a negative impact on these cells. Indeed, Nandrot et al. [29] reported that mice lacking ανβ5 integrin receptors fail to show the morning burst of phagocytic activity. During the aging process, these mice lose cone and rod photoreceptors more rapidly than wild-type mice, and the function of these cells is significantly impaired [29]. Thus, this study clearly indicated that the loss of the daily rhythm in RPE phagocytosis might reduce the photoreceptors’ viability during aging. A few years later, this group also reported that mice lacking MFG-E8 also do not show the phagocytic activity peak after light onset [30]. However, surprisingly, these mice did not show any significant difference during the aging process [90]. More recent investigations have also shown that mice lacking D2R or in which Bmal1 has been removed from the RPE do not show any negative phenotype in the photoreceptor function and RPE morphology, even during aging [31,42].
Finally, a recent investigation reported that in germline MT1 KO mice, there is a significant reduction in the number of photoreceptors at 18 months of age [5] and an increase in lipofuscin-like (i.e., autofluorescence) accumulation in the RPE [34]. Initially, we attributed this effect to the fact that removal of MT1 affected the time of the phagocytic peak [34]. However, recent data suggest that the presence of the phagocytic peak does not affect RPE or photoreceptor health and viability during aging [31,42]. Hence, we now believe that the direct action of MT1 signaling on the RPE and/or photoreceptor cells are responsible for the phenotype(s) observed in our previous study.
Thus, experimental evidence from different mouse models suggests that the loss of the diurnal phagocytic peak of the POS does not result in any deleterious effects in the retina and RPE during aging. As mentioned earlier, importantly, in all the mice models described above, the RPE total daily phagocytic activity was not different, although the peak was abolished. Thus, the increase in the basal phagocytic activity may compensate for the peak loss.

6. Conclusions

In conclusion, experimental evidence accumulated over the last fifty years has demonstrated that the daily burst in phagocytic activity by the RPE is present in several vertebrate species (diurnal and nocturnal) as well as in the rod and cone photoreceptors. The mechanisms controlling this burst are complex and involve the circadian clock in the RPE and a well-known circadian output of the retina (i.e., dopamine) (Figure 3). However, new experimental evidence suggests that such a peak is not important for the health of the RPE or the photoreceptors.

Author Contributions

K.B., V.G. and G.T. conceptualized and wrote the article. K.B. and G.T. design the figures. All authors have read and agreed to the published version of the manuscript.

Funding

SC1GM135112 (K.B.); R01EY026291; R21EY031821 (G.T.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors whose names are listed immediately below certify that they have NO affiliations with or involvement in any organization or entity with any financial interest (such as honoraria; educational grants; participation in speakers’ bureaus; membership, employment, consultancies, stock ownership, or other equity interest; and expert testimony or patent-licensing arrangements), or non-financial interest (such as personal or professional relationships, affiliations, knowledge or beliefs) in the subject matter or materials discussed in this manuscript. Author names: K.B., V.G. and G.T.

References

  1. Felder, M.-P.; Buhr, E.D.; Dkhissi-Benyahya, O.; Hicks, D.; Peirson, S.N.; Ribelayga, C.P.; Sandu, C.; Spessert, R.; Tosini, G. Ocular Clocks: Adapting Mechanisms for Eye Functions and Health. Investig. Opthalmology Vis. Sci. 2018, 59, 4856–4870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kondratov, R.V.; Kondratova, A.A.; Gorbacheva, V.Y.; Vykhovanets, O.V.; Antoch, M.P. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 2006, 20, 1868–1873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Storch, K.-F.; Paz, C.; Signorovitch, J.; Raviola, E.; Pawlyk, B.; Li, T.; Weitz, C.J. Intrinsic Circadian Clock of the Mammalian Retina: Importance for Retinal Processing of Visual Information. Cell 2007, 130, 730–741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sawant, O.B.; Horton, A.M.; Zucaro, O.F.; Chan, R.; Bonilha, V.L.; Samuels, I.S.; Rao, S. The Circadian Clock Gene Bmal1 Controls Thyroid Hormone-Mediated Spectral Identity and Cone Photoreceptor Function. Cell Rep. 2017, 21, 692–706. [Google Scholar] [CrossRef] [Green Version]
  5. Baba, K.; Pozdeyev, N.; Mazzoni, F.; Contreras-Alcantara, S.; Liu, C.; Kasamatsu, M.; Martinez-Merlos, T.; Strettoi, E.; Iuvone, P.M.; Tosini, G. Melatonin modulates visual function and cell viability in the mouse retina via the MT1 melatonin receptor. Proc. Natl. Acad. Sci. USA 2009, 106, 15043–15048. [Google Scholar] [CrossRef] [Green Version]
  6. Baba, K.; Piano, I.; Lyuboslavsky, P.; Chrenek, M.A.; Sellers, J.T.; Zhang, S.; Gargini, C.; He, L.; Tosini, G.; Iuvone, P.M. Removal of clock gene Bmal1 from the retina affects retinal development and accelerates cone photoreceptor degeneration during aging. Proc. Natl. Acad. Sci. USA 2018, 115, 13099–13104. [Google Scholar] [CrossRef] [Green Version]
  7. Baba, K.; Tosini, G. Aging Alters Circadian Rhythms in the Mouse Eye. J. Biol. Rhythm. 2018, 33, 441–445. [Google Scholar] [CrossRef]
  8. Gianesini, C.; Hiragaki, S.; Laurent, V.; Hicks, D.; Tosini, G. Cone Viability Is Affected by Disruption of Melatonin Receptors Signaling. Investig. Opthalmology Vis. Sci. 2016, 57, 94–104. [Google Scholar] [CrossRef]
  9. Ait-Hmyed, O.; Felder-Schmittbuhl, M.-P.; Garcia-Garrido, M.; Beck, S.C.; Seide, C.; Sothilingam, V.; Tanimoto, N.; Seeliger, W.M.; Bennis, M.; Hicks, D. Mice lacking Period 1 and Period 2 circadian clock genes exhibit blue cone photoreceptor defects. Eur. J. Neurosci. 2013, 37, 1048–1060. [Google Scholar] [CrossRef]
  10. Hakkari, O.A.; Acar, N.; Savier, E.; Spinnhirny, P.; Bennis, M.; Felder-Schmittbuhl, M.; Mendoza, J.; Hicks, D. Rev-Erbα modulates retinal visual processing and behavioral responses to light. FASEB J. 2016, 30, 3690–3701. [Google Scholar] [CrossRef] [Green Version]
  11. Kevany, B.M.; Palczewski, K. Phagocytosis of Retinal Rod and Cone Photoreceptors. Physiology 2010, 25, 8–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bok, D. The retinal pigment epithelium: A versatile partner in vision. J. Cell Sci. 1993, 1993, 189–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Nguyen-Legros, J.; Hicks, D. Renewal of photoreceptor outer segments and their phagocytosis by theretinal pigment epithelium. Adv. Virus Res. 2000, 196, 245–313. [Google Scholar] [CrossRef]
  14. LaVail, M.M. Chapter 44 Legacy of the RCS rat: Impact of a seminal study on retinal cell biology and retinal degenerative diseases. Prog. Brain Res. 2001, 131, 617–627. [Google Scholar] [CrossRef]
  15. Lavail, M.M. Interaction of environmental light and eye pigmentation with inherited retinal degenerations. Vis. Res. 1980, 20, 1173–1177. [Google Scholar] [CrossRef]
  16. LaVail, M.M. Rod Outer Segment Disk Shedding in Rat Retina: Relationship to Cyclic Lighting. Science 1976, 194, 1071–1074. [Google Scholar] [CrossRef]
  17. Grace, M.S.; Chiba, A.; Menaker, M. Circadian control of photoreceptor outer segment membrane turnover in mice genetically incapable of melatonin synthesis. Vis. Neurosci. 1999, 16, 909–918. [Google Scholar] [CrossRef]
  18. Besharse, J.C.; Hollyfield, J.G. Turnover of mouse photoreceptor outer segments in constant light and darkness. Investig. Ophthalmol. Vis. Sci. 1979, 18, 1019–1024. [Google Scholar]
  19. Teirstein, P.S.; Goldman, A.I.; O’Brien, P.J. Evidence for both local and central regulation of rat rod outer segment disc shedding. Investig. Ophthalmol. Vis. Sci. 1980, 19, 1268–1273. [Google Scholar]
  20. Terman, J.S.; Reme, C.E.; Terman, M. Rod outer segment disk shedding in rats with lesions of the suprachiasmatic nucleus. Brain Res. 1993, 605, 256–264. [Google Scholar] [CrossRef]
  21. Tosini, G.; Menaker, M. Circadian Rhythms in Cultured Mammalian Retina. Science 1996, 272, 419–421. [Google Scholar] [CrossRef] [PubMed]
  22. Ruan, G.-X.; Allen, G.C.; Yamazaki, S.; McMahon, D.G. An Autonomous Circadian Clock in the Inner Mouse Retina Regulated by Dopamine and GABA. PLoS Biol. 2008, 6, e249-18. [Google Scholar] [CrossRef] [PubMed]
  23. Tosini, G.; Davidson, A.J.; Fukuhara, C.; Kasamatsu, M.; Castanon-Cervantes, O. Localization of a circadian clock in mammalian photoreceptors. FASEB J. 2007, 21, 3866–3871. [Google Scholar] [CrossRef] [PubMed]
  24. Jaeger, C.; Sandu, C.; Malan, A.; Mellac, K.; Hicks, D.; Felder-Schmittbuhl, M. Circadian organization of the rodent retina involves strongly coupled, layer-specific oscillators. FASEB J. 2015, 29, 1493–1504. [Google Scholar] [CrossRef] [Green Version]
  25. Baba, K.; Sengupta, A.; Tosini, M.; Contreras-Alcantara, S.; Tosini, G. Circadian regulation of the PERIOD 2::LUCIFERASE bioluminescence rhythm in the mouse retinal pigment epithelium-choroid. Mol. Vis. 2010, 16, 2605. [Google Scholar]
  26. Baba, K.; DeBruyne, J.P.; Tosini, G. Dopamine 2 Receptor Activation Entrains Circadian Clocks in Mouse Retinal Pigment Epithelium. Sci. Rep. 2017, 7, 1–9. [Google Scholar]
  27. Bobu, C.; Craft, C.M.; Masson-Pevet, M.; Hicks, D. Photoreceptor Organization and Rhythmic Phagocytosis in the Nile RatArvicanthis Ansorgei: A Novel Diurnal Rodent Model for the Study of Cone Pathophysiology. Investig. Opthalmology Vis. Sci. 2006, 47, 3109–3118. [Google Scholar] [CrossRef] [Green Version]
  28. Krigel, A.; Felder-Schmittbuhl, M.-P.; Hicks, D. Circadian-clock driven cone-like photoreceptor phagocytosis in the neural retina leucine zipper gene knockout mouse. Mol. Vis. 2010, 16, 2873–2881. [Google Scholar] [PubMed]
  29. Nandrot, E.F.; Kim, Y.; Brodie, S.; Huang, X.; Sheppard, D.; Finnemann, S.C. Loss of Synchronized Retinal Phagocytosis and Age-related Blindness in Mice Lacking αvβ5 Integrin. J. Exp. Med. 2004, 200, 1539–1545. [Google Scholar] [CrossRef]
  30. Nandrot, E.; Anand, M.; Almeida, D.; Atabai, K.; Sheppard, D.; Finnemann, S.C. Essential role for MFG-E8 as ligand for vbeta5 integrin in diurnal retinal phagocytosis. Proc. Natl. Acad. Sci. USA 2007, 104, 12005–12010. [Google Scholar] [CrossRef] [Green Version]
  31. Goyal, V.; DeVera, C.; Laurent, V.; Sellers, J.; Chrenek, M.A.; Hicks, D.; Baba, K.; Iuvone, P.M.; Tosini, G. Dopamine 2 Receptor Signaling Controls the Daily Burst in Phagocytic Activity in the Mouse Retinal Pigment Epithelium. Investig. Opthalmology Vis. Sci. 2020, 61, 10. [Google Scholar] [CrossRef] [PubMed]
  32. Mao, Y.; Finnemann, S.C. Analysis of Photoreceptor Outer Segment Phagocytosis by RPE Cells in Culture. Program. Necrosis 2012, 935, 285–295. [Google Scholar] [CrossRef] [Green Version]
  33. Ruggiero, L.; Connor, M.P.; Chen, J.; Langen, R.; Finnemann, S.C. Diurnal, localized exposure of phosphatidylserine by rod outer segment tips in wild-type but not Itgb5-/- or Mfge8-/- mouse retina. Proc. Natl. Acad. Sci. USA 2012, 109, 8145–8148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Laurent, V.; Sengupta, A.; Sanchez, A.S.-B.; Hicks, D.; Tosini, G. Melatonin signaling affects the timing in the daily rhythm of phagocytic activity by the retinal pigment epithelium. Exp. Eye Res. 2017, 165, 90–95. [Google Scholar] [CrossRef]
  35. Gibbs, D.; Kitamoto, J.; Williams, D.S. Abnormal phagocytosis by retinal pigmented epithelium that lacks myosin VIIa, the Usher syndrome 1B protein. Proc. Natl. Acad. Sci. USA 2003, 100, 6481–6486. [Google Scholar] [CrossRef] [Green Version]
  36. Law, A.-L.; Ling, Q.; Hajjar, K.A.; Futter, C.E.; Greenwood, J.; Adamson, P.; Wavre-Shapton, S.T.; Moss, S.E.; Hayes, M.J. Annexin A2 Regulates Phagocytosis of Photoreceptor Outer Segments in the Mouse Retina. Mol. Biol. Cell 2009, 20, 3896–3904. [Google Scholar] [CrossRef] [Green Version]
  37. Mustafi, D.; Kevany, B.M.; Genoud, C.; Bai, X.; Palczewski, K. Photoreceptor phagocytosis is mediated by phosphoinositide signaling. FASEB J. 2013, 27, 4585–4595. [Google Scholar] [CrossRef] [Green Version]
  38. DeVera, C.; Tosini, G. Circadian analysis of the mouse retinal pigment epithelium transcriptome. Exp. Eye Res. 2020, 193, 107988. [Google Scholar] [CrossRef]
  39. Louer, E.M.; Günzel, D.; Rosenthal, R.; Carmone, C.; Yi, G.; Stunnenberg, H.G.; Hollander, A.I.D.; Deen, P.M. Differential day-night expression of tight junction components in murine retinal pigment epithelium. Exp. Eye Res. 2020, 193, 107985. [Google Scholar] [CrossRef]
  40. Chao, J.R.; Knight, K.; Engel, A.L.; Jankowski, C.; Wang, Y.; Manson, M.A.; Gu, H.; Djukovic, D.; Raftery, D.; Hurley, J.B.; et al. Human retinal pigment epithelial cells prefer proline as a nutrient and transport metabolic intermediates to the retinal side. J. Biol. Chem. 2017, 292, 12895–12905. [Google Scholar] [CrossRef] [Green Version]
  41. Milićević, N.; Hakkari, O.A.; Bagchi, U.; Sandu, C.; Jongejan, A.; Moerland, P.D.; Brink, J.B.T.; Hicks, D.; Bergen, A.A.; Felder-Schmittbuhl, M. Core circadian clock genes Per1 and Per2 regulate the rhythm in photoreceptor outer segment phagocytosis. FASEB J. 2021, 35, e21722. [Google Scholar] [CrossRef] [PubMed]
  42. DeVera, C.; Dixon, J.; Chrenek, M.A.; Baba, K.; Iuvone, P.M.; Tosini, G. The circadian clock in the retinal pigment epithelium controls the diurnal rhythm of phagocytic activity. bioRxiv 2020. [Google Scholar] [CrossRef]
  43. Nandrot, E.; Dufour, E.M.; Provost, A.C.; Péquignot, M.O.; Bonnel, S.; Gogat, K.; Marchant, D.; Rouillac, C.; de Condé, B.S.; Bihoreau, M.-T.; et al. Homozygous Deletion in the Coding Sequence of the c-mer Gene in RCS Rats Unravels General Mechanisms of Physiological Cell Adhesion and Apoptosis. Neurobiol. Dis. 2000, 7, 586–599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. D’Cruz, P.M.; Yasumura, D.; Weir, J.; Matthes, M.T.; Abderrahim, H.; Lavail, M.M.; Vollrath, D. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum. Mol. Genet. 2000, 9, 645–651. [Google Scholar] [CrossRef] [Green Version]
  45. Lu, Q.; Gore, M.; Zhang, Q.; Camenisch, T.; Boast, S.; Casagranda, F.; Lai, C.; Skinner, M.K.; Klein, R.; Matsushima, G.K.; et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 1999, 398, 723–728. [Google Scholar] [CrossRef]
  46. Brea-Fernandez, A.J.; Pomares, E.; Brion, M.J.; Marfany, G.; Blanco, M.J.; Sanchez-Salorio, M.; Gonzalez-Duarte, R.; Carracedo, A. Novel splice donor site mutation in MERTK gene associated with retinitis pigmentosa. Br. J. Ophthalmol. 2008, 92, 1419–1423. [Google Scholar] [CrossRef]
  47. Ksantini, M.; Lafont, E.; Bocquet, B.; Meunier, I.; Hamel, C.P. Homozygous Mutation in MERTK Causes Severe Autosomal Recessive Retinitis Pigmentosa. Eur. J. Ophthalmol. 2011, 22, 647–653. [Google Scholar] [CrossRef]
  48. Mackay, D.S.; Henderson, R.H.; Sergouniotis, P.I.; Li, Z.; Moradi, P.; Holder, G.E.; Waseem, N.; Bhattacharya, S.S.; Aldahmesh, M.A.; Alkuraya, F.S.; et al. Novel mutations in MERTK associated with childhood onset rod-cone dystrophy. Mol. Vis. 2010, 16, 369–377. [Google Scholar]
  49. McHenry, C.L.; Liu, Y.; Feng, W.; Nair, A.R.; Feathers, K.L.; Ding, X.; Gal, A.; Vollrath, U.; Sieving, P.A.; Thompson, D. MERTK arginine-844-cysteine in a patient with severe rod-cone dystrophy: Loss of mutant protein function in transfected cells. Investig. Opthalmology Vis. Sci. 2004, 45, 1456–1463. [Google Scholar] [CrossRef] [Green Version]
  50. Tschernutter, M.; Jenkins, S.A.; Waseem, N.H.; Saihan, Z.; E Holder, G.; Bird, A.C.; Bhattacharya, S.S.; Ali, R.R.; Webster, A.R. Clinical characterisation of a family with retinal dystrophy caused by mutation in the Mertk gene. Br. J. Ophthalmol. 2006, 90, 718–723. [Google Scholar] [CrossRef] [Green Version]
  51. Prasad, D.; Rothlin, C.V.; Burrola, P.; Burstyn-Cohen, T.; Lu, Q.; de Frutos, P.G.; Lemke, G. TAM receptor function in the retinal pigment epithelium. Mol. Cell. Neurosci. 2006, 33, 96–108. [Google Scholar] [CrossRef] [PubMed]
  52. Sugimoto, M.; Kondo, M.; Yasuma, T.; D’Alessandro-Gabazza, C.N.; Toda, M.; Imai, H.; Nakamura, M.; Gabazza, E.C. Increased expression of Protein S in eyes with diabetic retinopathy and diabetic macular edema. Sci. Rep. 2021, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  53. Adijanto, J.; Philp, N.J. Cultured primary human fetal retinal pigment epithelium (hfRPE) as a model for evaluating RPE metabolism. Exp. Eye Res. 2014, 126, 77–84. [Google Scholar] [CrossRef] [Green Version]
  54. Hu, J.; Bok, D. The use of cultured human fetal retinal pigment epithelium in studies of the classical retinoid visual cycle and retinoid-based disease processes. Exp. Eye Res. 2013, 126, 46–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Mayerson, P.L.; Hall, M.O.; Clark, V.; Abrams, T. An improved method for isolation and culture of rat retinal pigment epithelial cells. Investig. Ophthalmol. Vis. Sci. 1985, 26, 1599–1609. [Google Scholar]
  56. Sonoda, S.; Spee, C.; Barron, E.; Ryan, S.J.; Kannan, R.; Hinton, D.R. A protocol for the culture and differentiation of highly polarized human retinal pigment epithelial cells. Nat. Protoc. 2009, 4, 662–673. [Google Scholar] [CrossRef] [Green Version]
  57. Godino, R.F.; Garland, D.L.; Pierce, E. Isolation, culture and characterization of primary mouse RPE cells. Nat. Protoc. 2016, 11, 1206–1218. [Google Scholar] [CrossRef]
  58. Dunn, K.C.; Aotaki-Keen, A.E.; Putkey, F.R.; Hjelmeland, L.M. ARPE-19, A Human Retinal Pigment Epithelial Cell Line with Differentiated Properties. Exp. Eye Res. 1996, 62, 155–170. [Google Scholar] [CrossRef]
  59. Nabi, I.; Mathews, A.; Cohen-Gould, L.; Gundersen, D.; Rodriguez-Boulan, E. Immortalization of polarized rat retinal pigment epithelium. J. Cell Sci. 1993, 104, 37–49. [Google Scholar] [CrossRef]
  60. Kuznetsova, A.V.; Kurinov, A.M.; Aleksandrova, M.A. Cell Models to Study Regulation of Cell Transformation in Pathologies of Retinal Pigment Epithelium. J. Ophthalmol. 2014, 2014, 1–18. [Google Scholar] [CrossRef]
  61. Fronk, A.H.; Vargis, E. Methods for culturing retinal pigment epithelial cells: A review of current protocols and future recommendations. J. Tissue Eng. 2016, 7, 27493715. [Google Scholar] [CrossRef] [Green Version]
  62. Lakkaraju, A.; Umapathy, A.; Tan, L.X.; Daniele, L.; Philp, N.J.; Boesze-Battaglia, K.; Williams, D.S. The cell biology of the retinal pigment epithelium. Prog. Retin. Eye Res. 2020, 78, 100846. [Google Scholar] [CrossRef] [PubMed]
  63. Pavan, B.; Frigato, E.; Pozzati, S.; Prasad, P.D.; Bertolucci, C.; Biondi, C. Circadian clocks regulate adenylyl cyclase activity rhythms in human RPE cells. Biochem. Biophys. Res. Commun. 2006, 350, 169–173. [Google Scholar] [CrossRef]
  64. Yoshikawa, A.; Shimada, H.; Numazawa, K.; Sasaki, T.; Ikeda, M.; Kawashima, M.; Kato, N.; Tokunaga, K.; Ebisawa, T. Establishment of human cell lines showing circadian rhythms of bioluminescence. Neurosci. Lett. 2008, 446, 40–44. [Google Scholar] [CrossRef] [PubMed]
  65. Milićević, N.; Mazzaro, N.; De Bruin, I.; Wils, E.; Brink, J.T.; Asbroek, A.T.; Mendoza, J.; Bergen, A.; Felder-Schmittbuhl, M.-P. Rev-Erbα and Photoreceptor Outer Segments modulate the Circadian Clock in Retinal Pigment Epithelial Cells. Sci. Rep. 2019, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  66. Yoo, S.H.; Yamazaki, S.; Lowrey, P.L.; Shimomura, K.; Ko, C.H.; Buhr, E.D.; Siepka, S.M.; Hong, H.K.; Oh, W.J.; Yoo, O.J.; et al. PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc. Natl. Acad. Sci. USA 2004, 101, 5339–5346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Baba, K.; Davidson, A.J.; Tosini, G. Melatonin Entrains PER2::LUC Bioluminescence Circadian Rhythm in the Mouse Cornea. Invest. Ophthalmol. Vis. Sci. 2015, 56, 4753–4758. [Google Scholar] [CrossRef] [Green Version]
  68. Dunmire, J.; Dalvin, L.; Bouhenni, R.; Edward, D. Expression of Circadian Rhythm Genes in the Mouse Iris-Ciliary Body Complex. Investig. Ophthalmol. Vis. Sci. 2013, 54, 1994. [Google Scholar]
  69. Tsuchiya, S.; Buhr, E.D.; Higashide, T.; Sugiyama, K.; Van Gelder, R.N. Light entrainment of the murine intraocular pressure circadian rhythm utilizes non-local mechanisms. PLoS ONE 2017, 12, e0184790. [Google Scholar] [CrossRef]
  70. Evans, J.A.; Suen, T.-C.; Callif, B.L.; Mitchell, A.S.; Castanon-Cervantes, O.; Baker, K.M.; Kloehn, I.; Baba, K.; Teubner, B.J.W.; Ehlen, J.C.; et al. Shell neurons of the master circadian clock coordinate the phase of tissue clocks throughout the brain and body. BMC Biol. 2015, 13, 1–15. [Google Scholar] [CrossRef] [Green Version]
  71. Buhr, E.D.; Yue, W.W.S.; Ren, X.; Jiang, Z.; Liao, H.-W.R.; Mei, X.; Vemaraju, S.; Nguyen, M.-T.; Reed, R.R.; Lang, R.; et al. Neuropsin (OPN5)-mediated photoentrainment of local circadian oscillators in mammalian retina and cornea. Proc. Natl. Acad. Sci. USA 2015, 112, 13093–13098. [Google Scholar] [CrossRef] [Green Version]
  72. Ikarashi, R.; Akechi, H.; Kanda, Y.; Ahmad, A.; Takeuchi, K.; Morioka, E.; Sugiyama, T.; Ebisawa, T.; Ikeda, M.; Ikeda, M. Regulation of molecular clock oscillations and phagocytic activity via muscarinic Ca2+ signaling in human retinal pigment epithelial cells. Sci. Rep. 2017, 7, 44175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Besharse, J.C.; Dunis, D.A. Methoxyindoles and Photoreceptor Metabolism: Activation of Rod Shedding. Science 1983, 219, 1341–1343. [Google Scholar] [CrossRef] [PubMed]
  74. Reme, C.; Wirz-Justice, A.; Rhyner, A.; Hofmann, S. Circadian rhythm in the light response of rat retinal disk-shedding and autophagy. Brain Res. 1986, 369, 356–360. [Google Scholar] [CrossRef]
  75. Mariani, A.P.; Neff, N.H.; Hadjiconstantinou, M. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment decreases dopamine and increases lipofuscin in mouse retina. Neurosci. Lett. 1986, 72, 221–226. [Google Scholar] [CrossRef]
  76. Ginty, D.D.; Kornhauser, J.M.; Thompson, M.A.; Bading, H.; Mayo, K.E.; Takahashi, J.S.; Greenberg, M.E. Regulation of CREB Phosphorylation in the Suprachiasmatic Nucleus by Light and a Circadian Clock. Science 1993, 260, 238–241. [Google Scholar] [CrossRef]
  77. Ding, J.M.; Faiman, L.E.; Hurst, W.J.; Kuriashkina, L.R.; Gillette, M.U. Resetting the Biological Clock: Mediation of Nocturnal CREB Phosphorylation via Light, Glutamate, and Nitric Oxide. J. Neurosci. 1997, 17, 667–675. [Google Scholar] [CrossRef]
  78. Yagita, K.; Okamura, H. Forskolin induces circadian gene expression of rPer1, rPer2 and dbp in mammalian rat-1 fibroblasts. FEBS Lett. 1999, 465, 79–82. [Google Scholar] [CrossRef] [Green Version]
  79. Yan, Z.; Feng, J.; Fienberg, A.A.; Greengard, P. D2 dopamine receptors induce mitogen-activated protein kinase and cAMP response element-binding protein phosphorylation in neurons. Proc. Natl. Acad. Sci. USA 1999, 96, 11607–11612. [Google Scholar] [CrossRef] [Green Version]
  80. Choe, E.S.; Wang, J.Q. CaMKII regulates amphetamine-induced ERK1/2 phosphorylation in striatal neurons. Neuroreport 2002, 13, 1013–1016. [Google Scholar] [CrossRef]
  81. Illario, M.; Cavallo, A.L.; Bayer, K.U.; Di Matola, T.; Fenzi, G.; Rossi, G.; Vitale, M. Calcium/Calmodulin-dependent Protein Kinase II Binds to Raf-1 and Modulates Integrin-stimulated ERK Activation. J. Biol. Chem. 2003, 278, 45101–45108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Schmitt, J.M.; Wayman, G.A.; Nozaki, N.; Soderling, T.R. Calcium Activation of ERK Mediated by Calmodulin Kinase I. J. Biol. Chem. 2004, 279, 24064–24072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Obrietan, K.; Impey, S.; Storm, D.R. Light and circadian rhythmicity regulate MAP kinase activation in the suprachiasmatic nuclei. Nat. Neurosci. 1998, 1, 693–700. [Google Scholar] [CrossRef] [PubMed]
  84. Butcher, G.Q.; Lee, B.; Obrietan, K. Temporal Regulation of Light-Induced Extracellular Signal-Regulated Kinase Activation in the Suprachiasmatic Nucleus. J. Neurophysiol. 2003, 90, 3854–3863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Coogan, A.N.; Piggins, H.D. Circadian and photic regulation of phosphorylation of ERK1/2 and Elk-1 in the suprachiasmatic nuclei of the Syrian hamster. J. Neurosci. 2003, 23, 3085–3093. [Google Scholar] [CrossRef]
  86. Nakaya, M.; Sanada, K.; Fukada, Y. Spatial and temporal regulation of mitogen-activated protein kinase phosphorylation in the mouse suprachiasmatic nucleus. Biochem. Biophys. Res. Commun. 2003, 305, 494–501. [Google Scholar] [CrossRef]
  87. Baler, R.; Covington, S.; Klein, D.C. The Rat Arylalkylamine N-Acetyltransferase Gene Promoter: cAMP activation via A cAMP-responsive element-CCAAT complex. J. Biol. Chem. 1997, 272, 6979–6985. [Google Scholar] [CrossRef] [Green Version]
  88. Butcher, G.Q.; Lee, B.; Hsieh, F.; Obrietan, K. Light- and clock-dependent regulation of ribosomal S6 kinase activity in the suprachiasmatic nucleus. Eur. J. Neurosci. 2004, 19, 907–915. [Google Scholar] [CrossRef]
  89. Morioka, E.; Kanda, Y.; Koizumi, H.; Miyamoto, T.; Ikeda, M. Histamine Regulates Molecular Clock Oscillations in Human Retinal Pigment Epithelial Cells via H1 Receptors. Front. Endocrinol. 2018, 9, 108. [Google Scholar] [CrossRef] [Green Version]
  90. Yujnovsky, I.; Hirayama, J.; Doi, M.; Borrelli, E.; Sassone-Corsi, P. Signaling mediated by the dopamine D2 receptor potentiates circadian regulation by CLOCK:BMAL1. Proc. Natl. Acad. Sci. USA 2006, 103, 6386–6391. [Google Scholar] [CrossRef] [Green Version]
  91. Iuvone, P.M.; Galli, C.L.; Garrison-Gund, C.K.; Neff, N.H. Light Stimulates Tyrosine Hydroxylase Activity and Dopamine Synthesis in Retinal Amacrine Neurons. Science 1978, 202, 901–902. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of the molecular circadian clock. BMAL1:CLOCK heterodimer binds to the E-box present on the promoter region of the Per and Cry genes. Then, PERs, together with CRYs, inhibit their transcription by blocking the action of the BMAL1:CLOCK. The second feedback loop involves the transactivation of the Rev-Erb and Ror genes. REV-ERB and ROR compete for binding to RRE elements in the Bmal1 promoter, driving a daily rhythm of Bmal1 transcription. These feedback loops generate a 24-h rhythmic oscillation.
Figure 1. Schematic illustration of the molecular circadian clock. BMAL1:CLOCK heterodimer binds to the E-box present on the promoter region of the Per and Cry genes. Then, PERs, together with CRYs, inhibit their transcription by blocking the action of the BMAL1:CLOCK. The second feedback loop involves the transactivation of the Rev-Erb and Ror genes. REV-ERB and ROR compete for binding to RRE elements in the Bmal1 promoter, driving a daily rhythm of Bmal1 transcription. These feedback loops generate a 24-h rhythmic oscillation.
Ijms 23 02699 g001
Figure 3. Proposed mechanisms by which light regulates the burst in phagocytic activity after the onset of light. Photoreceptors (PRL) perceive light and then signal to dopaminergic neurons in the inner retina (INL) to release dopamine (DA) [91]. DA then diffuses within the retina and reaches the RPE. DA binds to D2R and activate integrin signaling thus stimulating phagocytic activity by RPE after the onset of light. Thus, DA signaling, via the D2R, conveys the light signal to the RPE that regulates the burst in phagocytic activity. The circadian clock in the RPE also controls the burst in phagocytic activity by controlling the expression of D2R.
Figure 3. Proposed mechanisms by which light regulates the burst in phagocytic activity after the onset of light. Photoreceptors (PRL) perceive light and then signal to dopaminergic neurons in the inner retina (INL) to release dopamine (DA) [91]. DA then diffuses within the retina and reaches the RPE. DA binds to D2R and activate integrin signaling thus stimulating phagocytic activity by RPE after the onset of light. Thus, DA signaling, via the D2R, conveys the light signal to the RPE that regulates the burst in phagocytic activity. The circadian clock in the RPE also controls the burst in phagocytic activity by controlling the expression of D2R.
Ijms 23 02699 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Baba, K.; Goyal, V.; Tosini, G. Circadian Regulation of Retinal Pigment Epithelium Function. Int. J. Mol. Sci. 2022, 23, 2699. https://doi.org/10.3390/ijms23052699

AMA Style

Baba K, Goyal V, Tosini G. Circadian Regulation of Retinal Pigment Epithelium Function. International Journal of Molecular Sciences. 2022; 23(5):2699. https://doi.org/10.3390/ijms23052699

Chicago/Turabian Style

Baba, Kenkichi, Varunika Goyal, and Gianluca Tosini. 2022. "Circadian Regulation of Retinal Pigment Epithelium Function" International Journal of Molecular Sciences 23, no. 5: 2699. https://doi.org/10.3390/ijms23052699

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop