Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2136 word(s) 2136 2022-03-02 02:59:02 |
2 format Meta information modification 2136 2022-03-21 02:09:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tosini, G. Circadian Regulation of Retinal Pigment Epithelium Function. Encyclopedia. Available online: https://encyclopedia.pub/entry/20730 (accessed on 03 July 2024).
Tosini G. Circadian Regulation of Retinal Pigment Epithelium Function. Encyclopedia. Available at: https://encyclopedia.pub/entry/20730. Accessed July 03, 2024.
Tosini, Gianluca. "Circadian Regulation of Retinal Pigment Epithelium Function" Encyclopedia, https://encyclopedia.pub/entry/20730 (accessed July 03, 2024).
Tosini, G. (2022, March 18). Circadian Regulation of Retinal Pigment Epithelium Function. In Encyclopedia. https://encyclopedia.pub/entry/20730
Tosini, Gianluca. "Circadian Regulation of Retinal Pigment Epithelium Function." Encyclopedia. Web. 18 March, 2022.
Circadian Regulation of Retinal Pigment Epithelium Function
Edit

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.

circadian melatonin dopamine RPE retina phagocytosis

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].
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.
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].

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.
  Function in RPE Animal Model Human Retinal Disease
MerTK receptor Outer segment binding & internalization RCS rat [43][44]
Merkd mouse [45]
Retinitis pigmentosa, rod-cone dystrophy [46][47][48][49][50]
Gas6, Protein S MerTK ligands Gas6 double KO and ProS1 [51] Diabetic Retinopathy and macular edema [52]
ανβ5 integrin receptor Outer segment binding,
Control the diurnal rhythm in peak of phagocytosis
β5−/− mouse [29] unknown
MFG-E8 ανβ5 integrin ligand
Control the diurnal rhythm in peak of phagocytosis
MFG-E8−/− mouse [30] unknown
Dopamine receptor 2 Controls the rhythm in RPE circadian clocks, light adaptation, peak of phagocytosis after light onset D2R KO mouse [31] unkown
Melatonin receptor 1 and 2 Control the timing of the peak of phagocytosis MT1 & MT2 KO mouse [34]  
RPE specific Bmal1 KO Control the diurnal rhythm in peak of phagocytosis RPEcre; Bmal1fl/fl [42] unknown
Per1/Per2 global KO Controls the amplitude of the peak of phagocytosis Per1−/−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].

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.
  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.
  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.
  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.
  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.
  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.
  7. Baba, K.; Tosini, G. Aging Alters Circadian Rhythms in the Mouse Eye. J. Biol. Rhythm. 2018, 33, 441–445.
  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.
  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.
  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.
  11. Kevany, B.M.; Palczewski, K. Phagocytosis of Retinal Rod and Cone Photoreceptors. Physiology 2010, 25, 8–15.
  12. Bok, D. The retinal pigment epithelium: A versatile partner in vision. J. Cell Sci. 1993, 1993, 189–195.
  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.
  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.
  15. Lavail, M.M. Interaction of environmental light and eye pigmentation with inherited retinal degenerations. Vis. Res. 1980, 20, 1173–1177.
  16. LaVail, M.M. Rod Outer Segment Disk Shedding in Rat Retina: Relationship to Cyclic Lighting. Science 1976, 194, 1071–1074.
  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.
  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.
  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.
  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.
  21. Tosini, G.; Menaker, M. Circadian Rhythms in Cultured Mammalian Retina. Science 1996, 272, 419–421.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  32. Mao, Y.; Finnemann, S.C. Analysis of Photoreceptor Outer Segment Phagocytosis by RPE Cells in Culture. Program. Necrosis 2012, 935, 285–295.
  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.
  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.
  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.
  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.
  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.
  38. DeVera, C.; Tosini, G. Circadian analysis of the mouse retinal pigment epithelium transcriptome. Exp. Eye Res. 2020, 193, 107988.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  57. Godino, R.F.; Garland, D.L.; Pierce, E. Isolation, culture and characterization of primary mouse RPE cells. Nat. Protoc. 2016, 11, 1206–1218.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
  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.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 368
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 21 Mar 2022
1000/1000
Video Production Service