Inactivation of the microRNA-183/96/182 cluster results in syndromic retinal degeneration.
Journal: 2013/April - Proceedings of the National Academy of Sciences of the United States of America
ISSN: 1091-6490
Abstract:
The microRNA-183/96/182 cluster is highly expressed in the retina and other sensory organs. To uncover its in vivo functions in the retina, we generated a knockout mouse model, designated "miR-183C(GT/GT)," using a gene-trap embryonic stem cell clone. We provide evidence that inactivation of the cluster results in early-onset and progressive synaptic defects of the photoreceptors, leading to abnormalities of scotopic and photopic electroretinograms with decreased b-wave amplitude as the primary defect and progressive retinal degeneration. In addition, inactivation of the miR-183/96/182 cluster resulted in global changes in retinal gene expression, with enrichment of genes important for synaptogenesis, synaptic transmission, photoreceptor morphogenesis, and phototransduction, suggesting that the miR-183/96/182 cluster plays important roles in postnatal functional differentiation and synaptic connectivity of photoreceptors.
Relations:
Content
Citations
(64)
References
(114)
Diseases
(2)
Conditions
(1)
Chemicals
(1)
Genes
(214)
Organisms
(3)
Processes
(5)
Anatomy
(4)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Proc Natl Acad Sci U S A 110(6): E507-E516

Inactivation of the microRNA<em>-183/96/182</em> cluster results in syndromic retinal degeneration

+4 authors

miR-183/96/182 Cluster Resides in the Intron of a Potentially Protein-Coding Gene.

The genomic features around miR-183/96/182 include two CpG islands ∼3.4–6.5 kb 5′ of premiR-183, the most 5′ miRNA of the cluster (Fig. 1A; and SI Appendix, Fig. S1 and Sequence (Seq.) S1) (http://genome.UCSC.edu/) (8). A cDNA clone, {"type":"entrez-nucleotide","attrs":{"text":"AK044220","term_id":"26090242","term_text":"AK044220"}}AK044220, extends ∼3.2–4.6 kb 5′ to premiR-183, encompassing the second CpG island (Fig. 1A and SI Appendix, Fig. S1). Multiple expressed sequences detected in gene-trap clones (27), including D016D06 (28, 29) [deposited by the German GeneTrap Consortium (GGTC) (http://tikus.gsf.de)], colocalize in {"type":"entrez-nucleotide","attrs":{"text":"AK044220","term_id":"26090242","term_text":"AK044220"}}AK044220 (Fig. 1A and SI Appendix, Fig. S1), suggesting a 5′ exon of the miR-183/96/182 gene. The region 3′ of miR-183/96/182 contains an EST clone, {"type":"entrez-nucleotide","attrs":{"text":"BB709579","term_id":"16062748","term_text":"BB709579"}}BB709579, ∼6 kb 3′ of premiR-182 (Fig. 1A), suggesting the miR-183/96/182 gene has a downstream exon. To confirm this model, we designed multiple primers complementary to the predicted exons to perform 5′ and 3′ RACE and RT-PCR using total RNA of the mouse retina and D016D06 ESCs followed by sequencing (see SI Appendix, Figs. S2 and S3 for details).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig01.jpg

Genomic structures of the miR-183/96/182 gene and the miR-183C allele of the gene-trap cell line D016D06. (A) Genomic features around the miR-183/96/182 gene compiled from the University of California, Santa Cruz Genome Browser (Mouse July 2007 Assembly). The blue arrow points to the gene-trap clone, D016D06. The open arrowhead points to the mouse cDNA clone, {"type":"entrez-nucleotide","attrs":{"text":"AK044220","term_id":"26090242","term_text":"AK044220"}}AK044220. tel, telomeric; cent, centromeric. (B) Genomic structure of the miR-183/96/182 gene. Blue boxes represent exons. The dotted blue lines indicate splicing events. pA, polyA signal. One-headed arrows mark locations and directions of exonic primers 2F, 2.1F, 3F, 3R, 5R, 5Rnest, and 6R. The numbers above the double-headed arrows denote the distances (in bp) between the vertical lines. The wavy end of Ex2 indicates that the 3′ end of this exon not yet fully defined. Alternative splicing events result in two forms of transcripts, transcripts I and II. Transcript II has four putative ORFs. In phase I of the sequence, two ORFs consist of 98 (P98-1) and 70 (P70-1) codons, respectively. Phases 2 and 3 each have an ORF of 63 codons (P63-2 and P63-3). (C) Genomic structure of the miR-183C allele of the gene-trap clone D016D06. The gene-trap construct was inserted in intron 1 of the miR-183/96/182 gene. Location and directions of primers, 4Fintron1, 5Fintron1, 353, 354, and 361 are labeled. LTR, long tandem repeat.

Our results showed that the miR-183/96/182 gene spans more than 15 kb, with at least two alternatively spliced transcripts (Fig. 1B). Both transcripts start from the nucleotide (nt) G, 3,943 nt 5′ to premiR-183 (−3943G) and use the same exon 1 (Ex1) (Fig. 1B and SI Appendix, Seq. S1). Transcript I is at least 979 nt and comprises Ex1 and exon 2 (Ex2), separated by a 3,213-bp intron. However, the 3′ RACE product detecting Ex2 stopped 197 nt 5′ to premiR-183 and did not extend beyond miR-183/96/182 (Fig. 1B and SI Appendix, Seq. S1). Thus, the 3′ end of transcript I is not yet fully determined. Transcript II is about 1,432 nt and consists of Ex1 and an alternatively spliced exon 2′ (Ex2′), which is 14 kb downstream of Ex1. Ex2′ is 986 bp in size with a polyA signal (Fig. 1B and SI Appendix, Seq. S1). The structure of transcript II clearly positions miR-183/96/182 in the intron of the miR-183/96/182 gene (Fig. 1B and SI Appendix, Seq. S1).

Transcript I has no ORF >30 codons, whereas transcript II has at least four ORFs, all of which reside in Ex 2′ (Fig. 1B and SI Appendix, Seqs. S2 and S3). In phase I of the sequence, two ORFs consist of 98 (P98-1) and 70 (P70-1) codons, respectively. Phases 2 and 3 each have an ORF of 63 codons (P63-2 and P63-3). None of the putative peptides contain known conserved functional domains; however, P98-1 encodes motifs found in proteins from multiple species (SI Appendix, Fig. S4). Thus, there is a possibility that the miR-183/96/182 gene encodes one or more peptides.

Characterization of the Gene-Trap Allele, miR-183C, and Production of miR-183C Mice.

Gene-trap clone D016D06 was derived from insertion of a retroviral construct, rFlpROSAβgeo, in 129S2 ESCs (Fig. 1 A and C) (http://tikus.gsf.de) (28, 29). The rFlpROSAβgeo construct carries a promoterless reporter gene, the β-geo cassette [an in-frame fusion of the β-galactosidase and neomycin resistance genes (30)], with a splicing acceptor (SA) immediately upstream and a polyA signal downstream of the β-geo cassette (Fig. 1C). To determine the location of the insertion of the construct in the “gene-trapped” allele, designated as “miR-183C,” we sequenced the product of genomic PCR using primers in intron 1 and in the gene-trap construct (Fig. 1C and SI Appendix, Seq. S1). Our results revealed that the gene-trap construct was inserted 1,848 bp downstream of Ex1 in intron 1 of the miR-183/96/182 gene. In the trapped allele, Ex1 of miR-183/96/182 gene spliced to β-geo, creating a fusion transcript (Fig. 1C) that stops after the polyA signal of the β-geo cassette, preventing transcription of miR-183/96/182. This result predicts that miR-183C mice would have complete loss of function of the miR-183/96/182 gene, whereas the β-geo cassette under the control of native regulatory elements should report endogenous expression pattern of the gene.

To test this model, we generated chimeric mice using the D016D06 ESCs (SI Appendix, Figs. S5–S7 and Table S1). Germline-transmitting F1 and F2 mice were produced. Genotypes of the F2 mice followed the expected Mendelian inheritance pattern (SI Appendix, Fig. S8). The miR-183C mice generally had decreased body weight (by ∼24%) and size (SI Appendix, Fig. S8); however, their overall survival and life span were not significantly different from their wild-type and heterozygous littermates.

miR-183/96/182 Gene Is Inactivated in the Retina and All Sensory Organs of miR-183C Mice.

Using RT-PCR (Fig. 2 A and C) and Northern blot analyses (Fig. 2B), we showed that both alternatively spliced transcripts (Fig. 2A) and three mature miRNAs of the miR-183/96/182 gene were undetectable in the retina of miR-183C mice (Fig. 2 B and C), whereas the Ex1/β-geo fusion transcript was present only in miR-183C but not in the wild-type retina (Fig. 2A). Similarly, all 3p miRNAs of the cluster also were abolished in the retina (SI Appendix, Fig. S9B).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig02.jpg

The miR-183/96/182 gene is inactivated in the retina and other sensory organs of miR-183C mice. (A) RT-PCR analysis of the transcripts of the miR-183/96/182 gene and the fusion transcript of the miR-183C allele. Transcript I was detected by RT-PCR using primers 2F/3R followed by nested PCR using 2.1F/3R-1. Transcript II was detected by RT-PCR using 2F/5R and nested PCR using 2.1F/5Rnest. The Ex1/β-geo fusion transcript of the miR-183C allele was detected by RT-PCR with primers 2F/254 and nested PCR with 2.1F/361. (B) Northern blot analysis. (CG) qRT-PCR analysis of the expression of members of the miR-183/96/182 cluster in the retina (C), IE (D), tongue epithelia (E), DRG (F), and OE (G). *P < 0.05; **P < 0.01; ***P < 0.001. miR-183C: n = 4; miR-183C: n = 4 (6 for DRG); miR-183C: n = 3 (4 for DRG).

To test whether the expression of miR-183/96/182 was affected in other sensory organs, we performed quantitative RT-PCR (qRT-PCR) analysis using RNA isolated from the inner ear (IE), tongue epithelia, dorsal root ganglions (DRGs), and olfactory epithelia (OE) (Fig. 2 DG) and found no detectable expression of miR-183/96/182 in any major sensory organs of miR-183C mice.

We conclude that the miR-183/96/182 gene was completely inactivated in miR-183C mice.

β-Geo Cassette Reports Endogenous Expression Patterns of the miR-183/96/182 Gene.

X-Gal staining in the adult miR-183C retina showed specific expression of β-gal in all photoreceptors in the outer nuclear layer (ONL) and in a subgroup of ganglion cells (Fig. 3 A–C), similar to the previously described expression pattern of miR-183/96/182 (5, 810). We also observed staining in the outer portion of the inner nuclear layer (INL); the punctate pattern suggested that the X-gal positive signals in the INL possibly were derived from synapses of the photoreceptors (SI Appendix, Fig. S10).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig03.jpg

Expression of the β-geo cassette reflects the endogenous expression patterns of the miR-183/96/182 gene in the retina. (AC) X-Gal staining of the retina. Red arrowheads in A and C show the expression in a subgroup of ganglion cells. OS, outer segment; IS, inner segment; IPL, inner plexiform layer; GCL, ganglion cell layer. (D) Expression of the β-geo cassette in photoreceptors of miR-183C mice has a circadian rhythm. X-Gal staining of retinal sections of miR-183C mice harvested every 4 h in a circadian cycle. (Scale bar: 20 μm.) Red dotted lines indicate unit squares used to measure optic density shown in E. (E) Relative optic density of X-Gal signals in the inner segment of photoreceptors in a unit square (illustrated in D). n = 3 at all time points. *P < 0.05; **P < 0.01 compared with CT1. (F) X-Gal staining of the retina of E14.5, E16.5, E18.5, P1, P2, P5, P10, and adult miR-183C mice. Arrows in P2 indicate positive signals in the outer neuroblast layer. The OPL started to form at P5. At this stage, ∼40% of photoreceptors are still in the INL (115) and are positive to X-Gal staining. INBL, inner neuroblast layer.

X-Gal staining of retina harvested every 4 h from adult miR-183C mice maintained in constant darkness showed that the intensity of X-Gal staining in photoreceptors followed a circadian rhythm, with peak intensity at Circadian Time (CT)1 and trough at CT13 (Fig. 3 D and E). This rhythm is in an approximately opposite phase to that we detected for miR-183/96/182 at the RNA level (8), likely reflecting the delay from transcription to the production of functional β-gal protein. Similar delays from transcription to protein expression have been reported in many other rhythmic genes (3133).

To demonstrate the developmental expression pattern, we performed X-Gal staining in the retinas of embryonic day (E)10.5, E14.5, E16.5, and E18.5 and postnatal day (P)1, P2, P5, and P10 heterozygous mice. The result showed that, similar to our previous qRT-PCR analysis on the mature miRNAs of the cluster (8), β-gal was not detectable in the embryonic stages or at P1 but started to express in the outer neuroblast layer (ONBL) at P2, was drastically up-regulated at P5, and was increased further at P10 and the adult stage (Fig. 3F). We conclude that the β-geo cassette of the miR-183C allele reports the endogenous expression pattern of the miR-183/96/182 gene in the retina developmentally, spatially, and temporally.

Also consistent with our previous results at the RNA level, X-Gal staining in other sensory organs of miR-183C mice showed specific expression in all sensory epithelia in the IE, OE, and the pheromone-sensing vomeronasal organ (VNO), papillae of tongue epithelia, and DRGs (Fig. 4; and SI Appendix, Figs. S11–S13). We also observed abundant expression in the olfactory bulbs (Fig. 4 C, a and SI Appendix, Fig. S14), possibly as a result of the expression in the axons and synaptic terminals of receptor neurons in the OE and VNO. Like many other retina-specific genes (34, 35), the miR-183/96/182 gene also is expressed in the pineal body (Fig. 4 C, b and SI Appendix, Fig. S14).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig04.jpg

Expression of the β-geo cassette reflects the endogenous expression pattern of the miR-183/96/182 cluster in other sensory and related organs. X-Gal staining of the IE (A a–c), OE and VNO (B), OB (C a), pineal body (C b), papillae of the tongue (D ad), and DRG (E a and b) of adult miR-183C mice.

Inactivation of the miR-183/96/182 Gene Results in Functional Defects in the Retina.

To assess retinal function, we performed scotopic (Fig. 5 A and B) and photopic (Fig. 5 C and D) electroretinograms (ERGs) on 5-wk-, 6-mo-, and 1-y-old miR-183C mice. The most prominent defect, decreased b-wave amplitude in miR-183C mice as compared with their age-matched, wild-type controls, was common to scotopic (Fig. 5 B, gi) and photopic ERGs (Fig. 5D) and was detectable in mice as early as 5 wk of age, at a time when the histology of the retina was normal (see below). The ERG abnormalities progressed with age, and photopic b-wave amplitudes were more severely affected than scotopic ERGs (Fig. 5 and SI Appendix, Fig. S15 and Table S2). At 1 y of age, the photopic b-wave amplitude of miR-183C mice was <10% that of wild-type littermate controls (Fig. 5D). The ERG b wave is derived mostly from activities of depolarizing bipolar cells (3639); defects in bipolar cells and photoreceptor synaptic transmission can produce defective b waves. Because miR-183/96/182 is expressed predominantly in photoreceptors, it is likely that the reduction in b-wave amplitude in miR-183C mice reflects defective synaptic transmission in rods and cones.

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig05.jpg

Inactivation of the miR-183/96/182 gene resulted in progressive ERG defects. (A and C) Representative scotopic (A) and photopic (C) ERG recordings of 5-wk-, 6-mo-, and 1-y-old mice. In A, blue, black, red, green, and violet lines represent recordings at 0, 10, 100, 1 k, and 10 k mcd⋅s⋅m, respectively. (B) Comparison of scotopic a- and b-wave amplitudes, latency, and implicit time of miR-183C and age-matched wild-type control mice. miR-183C: n = 12, 4, and 4 for 5-wk- (a, d, g, j), 6-mo- (b, e, h, k), and 1-y-old (c, f, i, l) mice, respectively; miR-183C: n = 10, 3, and 4 for 5-wk-, 6-mo-, and 1-y-old mice, respectively. *P < 0.05; **P < 0.01; ***P < 0.001 between miR-183C and age-matched wild-type controls. (D) Comparison of photopic a- and b-wave amplitudes, latency and implicit time of miR-183C and age-matched wild-type control mice. miR-183C: n = 3 at all ages; miR-183C: n = 4, 3, and 4 for 5-wk-, 6-mo-, and 1-y-old mice, respectively. Error bars indicate SEM. P < 0.05; P < 0.01; P < 0.001 between two age groups.

The photopic and scotopic a-wave amplitudes were normal in 5-wk-old miR-183C mice but decreased with age, and, as with the b wave, the photopic amplitude decreased more quickly than the scotopic amplitude (Fig. 5 B, ac and D): At 6 mo of age the photopic a-wave amplitude was reduced to 40% of control, and by 1 y it was <10% of control. The scotopic a-wave amplitude was normal until age 1 y, when it was reduced to 43–65% of control (Fig. 5D and SI Appendix, Table S2). The ERG a wave reflects the electrical response of photoreceptors to light (4042). These results suggest that in miR-183C mice the phototransduction pathways of rods and cones became significantly compromised with time, cones more so than rods.

ERG latency, the time from the onset of stimulus to the beginning of the a wave, and implicit time, the time from the onset of light stimulus to the peak of the b wave, of both scotopic and photopic ERGs were increased in miR-183C mice from age 6 mo, with more severe and persistent changes in photopic ERGs (Fig. 5 B and D and SI Appendix, Table S2), suggesting more severe synaptic transmission defects in the cone pathway.

ERG tests in heterozygous miR-183C mice at all ages showed no significant defects compared with their age-matched wild-type controls.

Inactivation of the miR-183/96/182 Gene Results in Progressive Retinal Degeneration.

At 5 wk of age, despite significant ERG abnormalities, the retinas of miR-183C mice were histologically indistinguishable from those of littermate controls (Fig. 6A). By age 6 mo, however, miR-183C mice showed retinal degeneration with decreased thickness of the ONL (Fig. 6B), which had progressed at 1 y of age (Fig. 6C). Intriguingly, the superior retina was more affected than the inferior retina. At age 6 mo, the number of nuclear layers of the ONL of the superior retina was decreased by ∼30%, whereas no significant changes were detected in the inferior retina (Fig. 6B). At age 1 y, ONL thickness was decreased in the entire eye, but the superior retina continued to be affected more severely (decreased by 40–52%) than the inferior retina (decreased by 30–43%) (Fig. 6C).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig06.jpg

Inactivation of the miR-183/96/182 gene resulted in progressive retinal degeneration and increased susceptibility to light damage. (AC) (Left) H&amp;E staining of sagittal sections of the retina of 5-wk- (A), 6-mo- (B), and 1-y-old (C) miR-183C and wild-type littermate control mice (miR-183C). (Right) Measurements of the thickness the ONL (Upper) and the number of nuclear layers of the ONL at different locations in reference to the optic nerve (Lower). Sup, superior; Inf, inferior. (D) H&amp;E staining (Left) and measurement (Right) of the retina of 6-mo-old miR-183C and wild-type littermate control mice 2 wk after a 2-h exposure to bright light. P < 0.06; *P < 0.05; **P < 0.01; ***P < 0.001.

Consistent with ERG results, heterozygous miR-183C mice showed no significant retinal degeneration compared with wild-type littermates at all ages tested.

Inactivation of the miR-183/96/182 Gene Results in Increased Susceptibility to Light Damage.

To determine if miR-183/96/182 influences the response of the retina to light damage, we exposed miR-183C mice and wild-type littermates to 10,000-lx cool, white fluorescent light for 2 h followed by 2 wk in the dark. Retinas of miR-183C mice showed increased light toxicity as compared with wild-type littermates (Fig. 6D and SI Appendix, Fig. S16). As with aging, the superior retina was more damaged than the inferior retina. The number of nuclear layers in ONL of the superior retina was decreased by 48–62% in miR-183C mice compared with their wild-type littermates; this decrease was similar to or worse than the extent of degeneration in 1-y-old miR-183C mice without light exposure (Fig. 6 C and D). The inferior retina had much less damage, suggesting that the superior retina is intrinsically more vulnerable to degenerative changes. This result is consistent with a recent report that knockdown of miR-183/96/182 in postmitotic photoreceptors resulted in increased susceptibility to light damage in the superior but not the inferior retina (26).

Inactivation of the miR-183/96/182 Gene Results in Photoreceptor Ribbon Synaptic Defects.

To identify target genes of miR-183/96/182 and to understand the mechanisms of functional defects in the retina of miR-183C mice, we performed gene-expression profiling using the Affymetrix GeneChip Mouse Genome 430A_2.0 and total RNA from 5-wk-old miR-183C mice, a time when there is no histological evidence of degeneration in the retina (SI Appendix, Fig. S17 and Tables S3–S9). A total of 1,341 genes showed significant differences in expression levels in miR-183C mice as compared with wild-type littermates (P < 0.05; fold change ≥1.2 folds) (SI Appendix, Tables S4–S6). Functional annotation analysis of the genes with altered expression showed significant enrichment for those involved in synaptogenesis (P = 3.2 × 10), synaptic contact (P = 3.9 × 10), and transmission of nerve impulses (P = 6.8 × 10) (SI Appendix, Table S7 A and B). In addition, key members of the classical complement system and Class I MHC (MHCI) molecules also were significantly enriched in the genes with increased expression (SI Appendix, Table S8). Members of the classical complement cascade and MHCI are expressed in retina and the central nervous system and play important roles in synaptic remodeling and connectivity (4349). These results are consistent with the ERG data indicating that inactivation of miR-183/96/182 gene results in abnormal photoreceptor synaptic transmission.

Neurotransmission of retinal photoreceptors is accomplished by a unique type of chemical synapse—the ribbon synapse—at their termini (50, 51). To evaluate the status of photoreceptor termini and ribbon synapses, we first performed immunofluorescence (IF) using synaptic markers. Ribeye/CtBP2, a major component of synaptic ribbons (52), normally has a horseshoe-shaped staining pattern in the wild-type photoreceptor termini (Fig. 7 A and C). However, Ribeye/CtBP2 staining became shorter and less defined, with dispersed immunoreactive puncta of variable size and less intensity in the miR-183C retina as early as age 5 wk (Fig. 7 A, B, E, and F), suggesting early-onset abnormalities of synaptic ribbons. This difference became more striking in 1-y-old animals (Fig. 7 C and D), suggesting the progression of synaptic defects. Consistently, peanut agglutinin (PNA) staining, which labels the inner and outer segment as well as pedicles of cone photoreceptors (53), showed that the number and size of cone pedicles apparently were decreased in 5-wk-old miR-183C mice (Fig. 7 E and F) and were reduced further in 1-y-old mice (SI Appendix, Fig. S18), suggesting defects in the maturation of photoreceptor termini.

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig07.jpg

Inactivation of the miR-183/96/182 gene resulted in morphological and molecular changes in the synaptic ribbons of photoreceptors in miR-183C mice. (AD) Confocal images of Ribeye IF in the retinas of 5-wk- (A and B) and 1-y-old (C and D) miR-183C mice (B and D) and their wild-type littermate controls (A and C). (E and F) Coimmunostaining of Ribeye (green) and PNA (red) in retinas of 5-wk-old miR-183C mice (F) and their wild-type littermate controls (E). (G) An electron micrograph of a ribbon synapse of an miR-183C mouse, showing a typical triad configuration of photoreceptor ribbon synapse. SR, synaptic ribbon; H, horizontal cell; B, bipolar cell. (H) Example of a 3D reconstruction of synaptic ribbons in the OPL of miR-183C mice (n = 2) and their wild-type littermates (miR-183C mice, n = 2), respectively. (Scale cube, 500 nm.) (I and J) Distribution of synaptic ribbons by size (I) and mean ribbon areas of miR-183C (red; n = 72) and miR-183C littermate controls (black; n = 49) (J). Data are shown as mean ± SEM; *P < 0.01.

To evaluate the photoreceptor synaptic structures directly, we performed EM of the retinas of 5-wk-old mice. Consistent with Ribeye/CtBP2 IF data, EM showed that, although synaptic ribbons were developed, ribbon size was decreased significantly in the miR-183C mice as compared with their wild-type littermates (Fig. 7 GJ).

Taken together, these observations support the notion that members of miR-183/96/182 modulate synaptic connections of photoreceptors to downstream neurons by directly and indirectly regulating a wide range of molecules involved in synaptogenesis and synaptic transmissions, contributing to retinal functional defects of miR-183C mice (see Fig. 9).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig09.jpg

Hypothetical model of in vivo functions of the miR-183/96/182 gene in the retina. Members of miR-183/96/182 directly and indirectly regulate multiple genes involved in photoreceptor maturation and synaptic connections with downstream bipolar cells. When the miR-183/96/182 gene is inactivated, the homeostasis of the photoreceptors is disrupted, resulting in defects in photoreceptors and their synaptic transmission, leading to progressive retinal dysfunction and degeneration.

Inactivation of the miR-183/96/182 Gene Disrupts Normal Expression of Photoreceptor Genes and Postnatal Development of Photoreceptors, Contributing to Retinal Degeneration.

Among the genes with abnormal expression are some known to be important in the phototransduction pathway and to be responsible for retinal diseases (SI Appendix, Table S9). By qRT-PCR analysis, we confirmed reduced expression of M-cone opsin, Opn1mw, and cone-specific arrestin, Arr3, by 2.54- and 2.74-fold, respectively (SI Appendix, Table S9; see also SI Appendix, Table S11B). IF of whole-mount retina and retinal sections validated that Opn1mw was decreased in the retina of 5-wk-old miR-183C mice (Fig. 8 A and B). Quantification of M opsin- and PNA-positive cones in the whole-mount retina showed that both the total number and the percentage of M opsin-positive cones in total cones (PNA-positive) were decreased significantly in the superior retina, by 49% and 26%, respectively (Fig. 8A), whereas no significant changes were seen in the total number of cones or the number of M opsin–positive cones in the inferior retina. At age 1 y M opsin was almost completely lost in miR-183C retina (Fig. 8C). These data suggest that M opsin-positive cones were the most affected photoreceptors in miR-183C mice and that this increased functional vulnerability of M opsin-positive cones leads to more severe and faster-progressing defects in photopic ERGs (Fig. 5) and to the uneven distribution of retinal degeneration in miR-183C mice (Fig. 6).

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655110fig08.jpg

Inactivation of the miR-183/96/182 gene resulted in a significant decrease of M opsin expression in the retina. PNA staining and co-IF of whole-mount retina (A) and retinal sections (B and C) of 5-wk- (A and B) and 1-y-old (C) miR-183C mice and their wild-type (miR-183C) littermate controls. The histograms at the bottom of A show the number of M opsin–positive (M opsin+) cones, the total number of cones per square micrometer, and the percentage of total cones that were M opsin positive in the superior (sup.) and inferior (inf.) retina.

Inactivation of the miR-183/96/182 Gene Also Results in Vestibular Dysfunction.

In addition to retinal defects, miR-183C mice exhibit circling behavior and an unstable gait (Movies S1 and S2). These phenotypic features are typical of defects in vestibular organs (54) and appeared as early as 4 wk of age. Because miR-183/96/182 is highly expressed in the vestibular organs (Fig. 4 A and C), this phenotype likely indicates defects in the vestibular organ and possibly in other sensory organs. In the current work we have focused on the retina.

Gene-Trap Cell Line and Mouse Production.

Gene-trap ESC clone D016D06, was obtained from the German Gene Trap Consortium (http://tikus.gsf.de/). Standard protocols of ESC culture were followed as described previously (108). Chimeric mice were produced at the Transgenic Core Facility of the University of Illinois at Chicago using standard protocols (109). See SI Appendix for more details of ESC characterization and mouse production. Mice were kept in 12-h light (<100 lx)12-h dark cycles with light on at 7:00 AM [zeitgeber time (ZT) 0] and off at 7:00 PM (ZT12). Animal care and husbandry followed National Institute of Health (NIH) and Association for Research in Vision and Ophthalmology (ARVO) guidelines. All protocols were approved by the Institutional Animal Care and Use Committee of Rush University Medical Center.

RNA Isolation and Northern Blot Analysis.

Protocols for RNA isolation and Northern blot analysis as described previously (8) were followed with modifications. See SI Appendix for more details.

ERG.

ERGs were carried out using a portable hmsERG machine (OccuScience). For scotopic ERG, we followed a protocol similar to that we described previously (110), with modifications. For photopic ERGs, a protocol similar to that previously described (111) was followed. See SI Appendix for more details.

Preparation of Cryoprotected Sections of Adult and Embryonic Mouse Retina.

A protocol similar to that described previously was followed (108). To collect embryonic retinas, we checked vaginal plugs in the females in the breeding cages every morning. When a female was plugged, we separated her from the male, recorded the date of plugging, and registered the predicted age of the embryo as 0.5 d postconception (d.p.c). We harvested the eyes at E14.5 and E16.6 and at 18.5 d.p.c. For postnatal age, we designated the day of birth as P1.

H&amp;E Staining and Measurement of Retinal Layers.

Standard H&amp;E staining was performed as described previously (108). Retinal thickness was measured using NIS-Elements BR software on a Nikon Eclipse 80i microscope. Moving outwards from the optic nerve, measurements of the OS, ONL, OPL, INL, and IPL were taken every 500 μm on live color images; the number of nuclear layers across the ONL also was counted. Statistical analysis was performed using two-tailed t tests.

X-Gal Staining.

X-Gal staining was performed as described previously (108). See SI Appendix for more details.

Antibodies, IF, and PNA Staining.

Antibodies against rhodopsin (Ab5417; AbCam), M opsin (AB5405; Millipore), S-opsin (AB5407; Millipore), Ribeye/CtBP2 (612044; BD Bioscience), PSD95 (75-028; Antibody Inc.), and synaptophysin (MAB5258; Millipore) were used as primary antibodies. Alexa Fluor-conjugated secondary antibodies were purchased from Invitrogen. IF was performed as described previously (112). Fluorescein and rhodamine-conjugated PNA was purchased from Vector Laboratories. For PNA staining on retinal sections, PNA (200 μg/mL) was incubated with sections for 1 h at room temperature followed by a 10-min postfixation. For whole-mount retinal IF, a protocol similar to that previously described (113) was followed. For quantification of cones in whole-mount retina, a protocol previously described by Ng et al. (95) was followed. See SI Appendix for more details.

EM.

Eye cups were fixed in 3% paraformaldehyde, 2.5% glutaraldehyde in 0.1 M phosphate buffer (pH 7.4) for 2 h at room temperature and then were processed for conventional EM as previously described (114). See SI Appendix for more details.

Gene-Expression Profiling, Data Analysis, and Confirmation Study.

GeneChip Mouse Genome 430A_2 Array (Affymetrix) was used for gene-expression profiling. The probe labeling, GeneChip hybridization, and data collection were performed at the Core Facility of Research Resource Center at the University of Illinois, Chicago. See SI Appendix for more details.

Light-Damage Experiment.

Mice were dark adapted overnight before the light-damage experiment. Pupils were dilated using 1% tropicamide solution and 2.5% phenylephrine (Bausch &amp; Lomb) for 15 min. Then mice were exposed for 2 h to 10,000-lx cool, white fluorescent light in cages lined with aluminum foil. After light exposure mice were kept in the dark for 2 wk before being killed for preparation of retinal sections.

Supplementary Material

Supporting Information:

Author Summary

Previously, we identified a cluster of miRNAs—small, noncoding, regulatory RNAs—that appears to be evolutionarily conserved and specifically expressed in sensory organs (1). Expression of this miR-183/96/182 cluster (also referred to as “miR-183/96/182”) is minimal in embryonic retina but increases dramatically after birth and peaks in adult retina with specific expression in photoreceptors (1). Reducing the expression of miR-183/96/182 in rod photoreceptors increased light damage to the mouse retina (2), but no histological or functional defects of the retina were observed under normal lighting conditions (2). Thus, in vivo functions of miR-183/96/182 in the retina remain uncertain.

We first dissected the genomic structure of the gene encoding miR-183/96/182 and showed that the miR-183/96/182 gene produces at least two primary RNA molecules, the transcripts (Fig. P1A). The structure of the second transcript clearly positions miR-183/96/182 in the intron of the gene. The second transcript has four predicted protein-coding regions, suggesting that the miR-183/96/182 gene may encode one or more peptides.

An external file that holds a picture, illustration, etc.
Object name is pnas.1212655S110fig01.jpg

(A) Genomic structures of the miR-183/96/182 gene. The dotted lines symbolize alternative splicing events. TSS, transcription start site. Ex, exon; pA, polyA signal. The numbers above the double-headed arrows denote sizes (in bp). (B and C) Functional defects detected by dark-adapted (B) and light-adapted (C) ERGs in 5-wk-old mutant (mut) mice. In B, blue, black, red, green, and violet lines represent recordings at 0, 10, 100, 1 k, and 10 k millicandela⋅s⋅m, respectively. (D) Retinal degeneration in 1-y-old mutant mice. (E) Hypothetical model of in vivo functions of miR-183/96/182 in the retina. When miR-183/96/182 is inactivated, homeostasis of photoreceptors is disrupted, resulting in defects in photoreceptors and their synaptic transmission, leading to retinal dysfunction and degeneration.

To study the functions of miR-183/96/182, we generated a mouse model in which the miR-183/96/182 gene is inactivated. Functionally, inactivation of the miR-183/96/182 gene resulted in early-onset, progressive electroretinogram (ERG) abnormalities. The most prominent defect, decreased b-wave amplitude, was common to both dark-adapted and light-adapted ERGs in the mutant mice (Fig. P1 B and C) and was detectable as early as 5 wk of age. The ERG b wave is derived mostly from activities of secondary neuron-bipolar cells; defects in bipolar cells and photoreceptor synaptic transmission can produce defective b waves (3). Because miR-183/96/182 is expressed predominantly in photoreceptors, reduction in b-wave amplitude likely reflects synaptic transmission defects in photoreceptors of miR-183/96/182–mutant mice. The amplitude, latency, and implicit time of the a wave, which reflects the activities of photoreceptors, appeared normal in 5-wk-old mutant mice but became defective with age. In all parameters, light-adapted ERG was more affected than dark-adapted ERG, suggesting more severe defects in the cone pathways.

Histologically, despite significant ERG abnormalities, retinas of 5-wk-old mutant mice were indistinguishable from littermate controls. By 6 mo, however, the mutant mice showed significant retinal degeneration, which progressed at 1 y of age (Fig. P1D). Intriguingly, the superior retina, which is enriched with red/green cones, was more affected than the inferior retina, which is enriched with blue cones.

A light-damage experiment showed significantly increased degeneration in the retinas of the mutant mice as compared with wild-type littermates after a 2-h exposure to 10,000-lx cool, white fluorescent light. In this experiment, as with aging, the superior retina was more damaged than the inferior retina, consistent with a recent report in a miR-183/96/182 knockdown mouse model (2).

Gene-expression profiling revealed global gene-expression changes in retinas of 5-wk-old miR-183/96/182–mutant mice. Genes important for synaptogenesis, nerve-impulse transmission, and synaptic remodeling were enriched significantly in the dysregulated genes, consistent with ERG data indicating abnormal synaptic transmission of photoreceptors. Electron microscopy and immunostaining with synaptic markers confirmed that the sizes of synaptic ribbons were decreased significantly in the mutant mice as early as age 5 wk.

Many genes known to be involved in phototransduction, a process by which light signals are transformed into neuroelectrical impulses in photoreceptors, also were dysregulated. We confirmed that the expression of red/green opsin was decreased significantly. Quantification of red/green opsin-positive and total cones in the whole-mount retina showed that both the number and the percentage of red/green opsin-positive cones were decreased significantly in the superior retina, but in the inferior retina the total number of cones and the number of red/green opsin-positive cones did not change significantly, suggesting that red/green cones were the most affected photoreceptors and that the greater vulnerability of red/green cones contributed to more severe defects in photopic ERGs and more degeneration in the superior retina in miR-183/96/182–mutant mice.

In mouse, both rod and cone photoreceptor differentiation and synaptogenesis occur postnatally starting from postnatal day 4–5 (4), coincident with developmental up-regulation of the miR-183/96/182 gene (1). Our data suggest that miR-183/96/182 is required for postnatal differentiation of photoreceptors and establishment of fully functional synaptic connections with their downstream neurons by directly and indirectly regulating genes important for synaptic functions and the phototransduction pathway. Inactivation of the miR-183/96/182 gene disrupts the homeostasis and functions of photoreceptors, leading to progressive retinal dysfunction and degeneration (Fig. P1E).

In addition to retinal defects, miR-183/96/182–mutant mice exhibited a circling behavior typical of defects in vestibular organs, suggesting the presence of syndromic sensory defects and that the requirement for the miR-183/96/182 gene for normal functions of sensory organs other than the retina. Studies on genetic variations around miR-183/96/182 gene are warranted in patients with progressive, sensory syndromic retinal dysfunction and other neurological conditions.

Departments of Pharmacology,
Ophthalmology, and
Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612;
Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, 21287;
Biosciences Division, Argonne National Laboratory, Lemont, IL, 60439; and
McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205
To whom correspondence should be addressed. E-mail: ude.hsur@ux_nibnuhs.
Edited by Jeremy Nathans, The Johns Hopkins University, Baltimore, MD, and approved December 21, 2012 (received for review July 31, 2012)

Author contributions: D.V., D.J.Z., D.A.N., and S.X. designed research; S.L., C.E.H., N.J.C., K.J.W., C.C., S.T., B.K., P.D.W., D.A.N., and S.X. performed research; S.L., C.E.H., N.J.C., K.J.W., C.C., P.E.L., D.A.N., and S.X. analyzed data; and S.X. wrote the paper.

Edited by Jeremy Nathans, The Johns Hopkins University, Baltimore, MD, and approved December 21, 2012 (received for review July 31, 2012)

MicroRNAs (miRNAs) are small, endogenous, noncoding, regulatory RNAs and represent a newly recognized level of gene-expression regulation (14). miRNAs have unique expression profiles in the developing and adult retina and are involved in normal development and functions of the retina in all species studied so far (512). miRNAs are dysregulated in the retina of retinal degenerative mouse models, suggesting their potential involvement in retinal degeneration (13, 14). Conditional inactivation of dicer, an RNase III endonuclease required for miRNA maturation in cytosol (15), in the mouse retina resulted in alteration of retinal differentiation and optic-cup patterning, increased cell death, and disorganization of axons of retinal ganglion cells (1619), suggesting that miRNAs are important for normal development and functions of the mammalian retina. However, in vivo functions of individual miRNAs in the retina still are largely unknown.

Previously, we identified a highly conserved, intergenic, sensory organ-specific, paralogous miRNA cluster, the miR-183/96/182 cluster (hereafter, miR-183/96/182), contained within an ∼4-kb genomic segment on mouse chr6qA3.3 (8, 9). In the adult retina, miR-183/96/182 is expressed specifically in all photoreceptors and in the inner nuclear layer (8, 10). Developmentally, its expression is minimal in the embryonic retina but increases dramatically after birth and peaks in the adult retina, suggesting a role for miR-183/96/182 in maturation and normal functioning of the adult retina (8, 9). Additionally, expression of miR-183/96/182 has a diurnal pattern, suggesting a potential role in rhythmic functions of the retina (8, 9). Recently, miR-183/96/182 also was shown to be light responsive, independent of the circadian cycle (20). Targeted deletion of miR-182 alone in mouse did not result in a discernible phenotype, suggesting functional compensation by miR-183 and miR-96 (21). Point mutations of miR-96 were reported to result in progressive, nonsyndromic hearing loss in both human (22) and mouse (23); however, there was no apparent retinal phenotype, an observation that suggests miR-96 plays a major role in the inner ear but not in the retina (2225). Finally, a recent report showed that knockdown of miR-183/96/182 in postmitotic rod photoreceptors in a miRNA-sponge transgenic mouse model resulted in increased susceptibility to light damage in the retina (26); however, no histological or functional defects of the retina were observed under normal lighting conditions (26). Thus, in vivo functions of miR-183/96/182 in the retina remain uncertain.

To search for the in vivo functions of miR-183/96/182, we first dissected the genomic structure of the gene encoding miR-183/96/182 (hereafter referred to as “the miR-183/96/182 gene”) and characterized a gene-trap embryonic stem cell (ESC) clone (2729) in which the gene-trap construct was inserted downstream of the first exon of the miR-183/96/182 gene, designated as “miR-183C allele.” Using this ESC clone, we generated a mouse model, designated as “miR-183C,” in which the miR-183/96/182 gene is inactivated, and the β-geo cassette in the gene-trap construct reliably mirrors the endogenous expression patterns of miR-183/96/182.

Click here to view.

Acknowledgments

We thank Dr. Jeremy Nathans for his comments on the manuscript and Dr. Yanshu Wang for technical consultation on PNA staining and whole-mount retinal IF. This work was supported by a grant from the Lincy Foundation (to S.X.).

Acknowledgments

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Data deposition: The sequences reported in this paper have been deposited in the GenBank database (accession nos. {"type":"entrez-nucleotide","attrs":{"text":"KC408930","term_id":"474854481","term_text":"KC408930"}}KC408930 and {"type":"entrez-nucleotide","attrs":{"text":"KC408931","term_id":"474854482","term_text":"KC408931"}}KC408931).

See Author Summary on page 1990 (volume 110, number 6).

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1212655110/-/DCSupplemental.

Footnotes
1. Xu S, Witmer PD, Lumayag S, Kovacs B, Valle D. MicroRNA (miRNA) transcriptome of mouse retina and identification of a sensory organ-specific miRNA cluster. J Biol Chem. 2007;282(34):25053–25066. [PubMed] [Google Scholar]
2. Zhu Q, et al. Sponge transgenic mouse model reveals important roles for the microRNA-183 (miR-183)/96/182 cluster in postmitotic photoreceptors of the retina. J Biol Chem. 2011;286(36):31749–31760.[PMC free article] [PubMed] [Google Scholar]
3. McCall MA, Gregg RG. Comparisons of structural and functional abnormalities in mouse b-wave mutants. J Physiol. 2008;586(Pt 18):4385–4392.[PMC free article] [PubMed] [Google Scholar]
4. Rich KA, Zhan Y, Blanks JC. Migration and synaptogenesis of cone photoreceptors in the developing mouse retina. J Comp Neurol. 1997;388(1):47–63. [PubMed] [Google Scholar]

References

  • 1. Ambros VThe functions of animal microRNAs. Nature. 2004;431(7006):350–355.[PubMed][Google Scholar]
  • 2. Bartel DPMicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–297.[PubMed][Google Scholar]
  • 3. Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell. 1993;75(5):855–862.[PubMed]
  • 4. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–854.[PubMed]
  • 5. Ryan DG, Oliveira-Fernandes M, Lavker RMMicroRNAs of the mammalian eye display distinct and overlapping tissue specificity. Mol Vis. 2006;12:1175–1184.[PubMed][Google Scholar]
  • 6. Li X, Carthew RWA microRNA mediates EGF receptor signaling and promotes photoreceptor differentiation in the Drosophila eye. Cell. 2005;123(7):1267–1277.[PubMed][Google Scholar]
  • 7. Wienholds E, et al MicroRNA expression in zebrafish embryonic development. Science. 2005;309(5732):310–311.[PubMed][Google Scholar]
  • 8. Xu S, Witmer PD, Lumayag S, Kovacs B, Valle DMicroRNA (miRNA) transcriptome of mouse retina and identification of a sensory organ-specific miRNA cluster. J Biol Chem. 2007;282(34):25053–25066.[PubMed][Google Scholar]
  • 9. Xu SmicroRNA expression in the eyes and their significance in relation to functions. Prog Retin Eye Res. 2009;28(2):87–116.[PubMed][Google Scholar]
  • 10. Karali M, Peluso I, Marigo V, Banfi SIdentification and characterization of microRNAs expressed in the mouse eye. Invest Ophthalmol Vis Sci. 2007;48(2):509–515.[PubMed][Google Scholar]
  • 11. Arora A, McKay GJ, Simpson DAPrediction and verification of miRNA expression in human and rat retinas. Invest Ophthalmol Vis Sci. 2007;48(9):3962–3967.[PubMed][Google Scholar]
  • 12. Hackler L, Jr, Wan J, Swaroop A, Qian J, Zack DJMicroRNA profile of the developing mouse retina. Invest Ophthalmol Vis Sci. 2010;51(4):1823–1831.[Google Scholar]
  • 13. Loscher CJ, et al Altered retinal microRNA expression profile in a mouse model of retinitis pigmentosa. Genome Biol. 2007;8(11):R248.[Google Scholar]
  • 14. Loscher CJ, et al A common microRNA signature in mouse models of retinal degeneration. Exp Eye Res. 2008;87(6):529–534.[Google Scholar]
  • 15. Lee YS, et al Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways. Cell. 2004;117(1):69–81.[PubMed][Google Scholar]
  • 16. Pinter R, Hindges RPerturbations of microRNA function in mouse dicer mutants produce retinal defects and lead to aberrant axon pathfinding at the optic chiasm. PLoS ONE. 2010;5(4):e10021.[Google Scholar]
  • 17. Davis N, Mor E, Ashery-Padan RRoles for Dicer1 in the patterning and differentiation of the optic cup neuroepithelium. Development. 2011;138(1):127–138.[PubMed][Google Scholar]
  • 18. Georgi SA, Reh TADicer is required for the transition from early to late progenitor state in the developing mouse retina. J Neurosci. 2010;30(11):4048–4061.[Google Scholar]
  • 19. Damiani D, et al Dicer inactivation leads to progressive functional and structural degeneration of the mouse retina. J Neurosci. 2008;28(19):4878–4887.[Google Scholar]
  • 20. Krol J, et al Characterizing light-regulated retinal microRNAs reveals rapid turnover as a common property of neuronal microRNAs. Cell. 2010;141(4):618–631.[PubMed][Google Scholar]
  • 21. Jin ZB, et al Targeted deletion of miR-182, an abundant retinal microRNA. Mol Vis. 2009;15:523–533.[Google Scholar]
  • 22. Mencía A, et al Mutations in the seed region of human miR-96 are responsible for nonsyndromic progressive hearing loss. Nat Genet. 2009;41(5):609–613.[PubMed][Google Scholar]
  • 23. Lewis MA, et al An ENU-induced mutation of miR-96 associated with progressive hearing loss in mice. Nat Genet. 2009;41(5):614–618.[Google Scholar]
  • 24. Kuhn S, et al miR-96 regulates the progression of differentiation in mammalian cochlear inner and outer hair cells. Proc Natl Acad Sci USA. 2011;108(6):2355–2360.[Google Scholar]
  • 25. Li H, Kloosterman W, Fekete DMMicroRNA-183 family members regulate sensorineural fates in the inner ear. J Neurosci. 2010;30(9):3254–3263.[Google Scholar]
  • 26. Zhu Q, et al Sponge transgenic mouse model reveals important roles for the microRNA-183 (miR-183)/96/182 cluster in postmitotic photoreceptors of the retina. J Biol Chem. 2011;286(36):31749–31760.[Google Scholar]
  • 27. Stanford WL, Cohn JB, Cordes SPGene-trap mutagenesis: Past, present and beyond. Nat Rev Genet. 2001;2(10):756–768.[PubMed][Google Scholar]
  • 28. Hansen J, et al A large-scale, gene-driven mutagenesis approach for the functional analysis of the mouse genome. Proc Natl Acad Sci USA. 2003;100(17):9918–9922.[Google Scholar]
  • 29. Schnütgen F, et al Genomewide production of multipurpose alleles for the functional analysis of the mouse genome. Proc Natl Acad Sci USA. 2005;102(20):7221–7226.[Google Scholar]
  • 30. Friedrich G, Soriano PPromoter traps in embryonic stem cells: A genetic screen to identify and mutate developmental genes in mice. Genes Dev. 1991;5(9):1513–1523.[PubMed][Google Scholar]
  • 31. Antoch MP, et al Functional identification of the mouse circadian Clock gene by transgenic BAC rescue. Cell. 1997;89(4):655–667.[Google Scholar]
  • 32. King DP, et al Positional cloning of the mouse circadian clock gene. Cell. 1997;89(4):641–653.[Google Scholar]
  • 33. Hastings MH, Herzog EDClock genes, oscillators, and cellular networks in the suprachiasmatic nuclei. J Biol Rhythms. 2004;19(5):400–413.[PubMed][Google Scholar]
  • 34. Lolley RN, Craft CM, Lee RHPhotoreceptors of the retina and pinealocytes of the pineal gland share common components of signal transduction. Neurochem Res. 1992;17(1):81–89.[PubMed][Google Scholar]
  • 35. Blackshaw S, Snyder SHDevelopmental expression pattern of phototransduction components in mammalian pineal implies a light-sensing function. J Neurosci. 1997;17(21):8074–8082.[Google Scholar]
  • 36. Robson JG, Frishman LJResponse linearity and kinetics of the cat retina: The bipolar cell component of the dark-adapted electroretinogram. Vis Neurosci. 1995;12(5):837–850.[PubMed][Google Scholar]
  • 37. Stockton RA, Slaughter MM. B-wave of the electroretinogram. A reflection of ON bipolar cell activity. J Gen Physiol. 1989;93(1):101–122.
  • 38. Hood DC, Birch DGBeta wave of the scotopic (rod) electroretinogram as a measure of the activity of human on-bipolar cells. J Opt Soc Am A Opt Image Sci Vis. 1996;13(3):623–633.[PubMed][Google Scholar]
  • 39. McCall MA, Gregg RGComparisons of structural and functional abnormalities in mouse b-wave mutants. J Physiol. 2008;586(Pt 18):4385–4392.[Google Scholar]
  • 40. Penn RD, Hagins WASignal transmission along retinal rods and the origin of the electroretinographic a-wave. Nature. 1969;223(5202):201–204.[PubMed][Google Scholar]
  • 41. Peachey NS, Goto Y, Quiambao AB, al-Ubaidi MRFunctional consequences of oncogene-induced photoreceptor degeneration in transgenic mice. Vis Neurosci. 1995;12(3):513–522.[PubMed][Google Scholar]
  • 42. Lyubarsky AL, Pugh EN., Jr Recovery phase of the murine rod photoresponse reconstructed from electroretinographic recordings. J Neurosci. 1996;16(2):563–571.
  • 43. Stevens B, et al The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131(6):1164–1178.[PubMed][Google Scholar]
  • 44. Perry VH, O’Connor VC1q: The perfect complement for a synaptic feast? Nat Rev Neurosci. 2008;9(11):807–811.[PubMed][Google Scholar]
  • 45. Corriveau RA, Huh GS, Shatz CJRegulation of class I MHC gene expression in the developing and mature CNS by neural activity. Neuron. 1998;21(3):505–520.[PubMed][Google Scholar]
  • 46. Huh GS, et al Functional requirement for class I MHC in CNS development and plasticity. Science. 2000;290(5499):2155–2159.[Google Scholar]
  • 47. Shatz CJMHC class I: An unexpected role in neuronal plasticity. Neuron. 2009;64(1):40–45.[Google Scholar]
  • 48. Boulanger LMImmune proteins in brain development and synaptic plasticity. Neuron. 2009;64(1):93–109.[PubMed][Google Scholar]
  • 49. Datwani A, et al Classical MHCI molecules regulate retinogeniculate refinement and limit ocular dominance plasticity. Neuron. 2009;64(4):463–470.[Google Scholar]
  • 50. Dowling JE, Boycott BBOrganization of the primate retina: Electron microscopy. Proc R Soc Lond B Biol Sci. 1966;166(2):80–111.[PubMed][Google Scholar]
  • 51. Cohen AIThe ultrastructure of the rods of the mouse retina. Am J Anat. 1960;107:23–48.[PubMed][Google Scholar]
  • 52. Schmitz F, Königstorfer A, Südhof TCRIBEYE, a component of synaptic ribbons: A protein’s journey through evolution provides insight into synaptic ribbon function. Neuron. 2000;28(3):857–872.[PubMed][Google Scholar]
  • 53. Blanks JC, Johnson LV. Specific binding of peanut lectin to a class of retinal photoreceptor cells. A species comparison. Invest Ophthalmol Vis Sci. 1984;25(5):546–557.[PubMed]
  • 54. Wenngren BI, Anniko MVestibular hair cell pathology in the dancer mouse mutant. Acta Otolaryngol. 1989;107(3-4):182–190.[PubMed][Google Scholar]
  • 55. Marson A, et al Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell. 2008;134(3):521–533.[Google Scholar]
  • 56. Ozsolak F, et al Chromatin structure analyses identify miRNA promoters. Genes Dev. 2008;22(22):3172–3183.[Google Scholar]
  • 57. Copenhagen DR, Jahr CERelease of endogenous excitatory amino acids from turtle photoreceptors. Nature. 1989;341(6242):536–539.[PubMed][Google Scholar]
  • 58. Gaal L, et al Postsynaptic response kinetics are controlled by a glutamate transporter at cone photoreceptors. J Neurophysiol. 1998;79(1):190–196.[PubMed][Google Scholar]
  • 59. Hasegawa J, Obara T, Tanaka K, Tachibana MHigh-density presynaptic transporters are required for glutamate removal from the first visual synapse. Neuron. 2006;50(1):63–74.[PubMed][Google Scholar]
  • 60. Zimmerman G, Soreq HTermination and beyond: Acetylcholinesterase as a modulator of synaptic transmission. Cell Tissue Res. 2006;326(2):655–669.[PubMed][Google Scholar]
  • 61. Grisaru D, Sternfeld M, Eldor A, Glick D, Soreq HStructural roles of acetylcholinesterase variants in biology and pathology. Eur J Biochem. 1999;264(3):672–686.[PubMed][Google Scholar]
  • 62. Friese MB, Blagden CS, Burden SJSynaptic differentiation is defective in mice lacking acetylcholine receptor beta-subunit tyrosine phosphorylation. Development. 2007;134(23):4167–4176.[PubMed][Google Scholar]
  • 63. Cao H, Chen J, Krueger EW, McNiven MASRC-mediated phosphorylation of dynamin and cortactin regulates the “constitutive” endocytosis of transferrin. Mol Cell Biol. 2010;30(3):781–792.[Google Scholar]
  • 64. Kruchten AE, Krueger EW, Wang Y, McNiven MADistinct phospho-forms of cortactin differentially regulate actin polymerization and focal adhesions. Am J Physiol Cell Physiol. 2008;295(5):C1113–C1122.[Google Scholar]
  • 65. Krueger EW, Orth JD, Cao H, McNiven MAA dynamin-cortactin-Arp2/3 complex mediates actin reorganization in growth factor-stimulated cells. Mol Biol Cell. 2003;14(3):1085–1096.[Google Scholar]
  • 66. Gray NW, Kruchten AE, Chen J, McNiven MAA dynamin-3 spliced variant modulates the actin/cortactin-dependent morphogenesis of dendritic spines. J Cell Sci. 2005;118(Pt 6):1279–1290.[PubMed][Google Scholar]
  • 67. Knöll B, Drescher USrc family kinases are involved in EphA receptor-mediated retinal axon guidance. J Neurosci. 2004;24(28):6248–6257.[Google Scholar]
  • 68. Hindges R, McLaughlin T, Genoud N, Henkemeyer M, O’Leary DDEphB forward signaling controls directional branch extension and arborization required for dorsal-ventral retinotopic mapping. Neuron. 2002;35(3):475–487.[PubMed][Google Scholar]
  • 69. Liu X, Hawkes E, Ishimaru T, Tran T, Sretavan DWEphB3: An endogenous mediator of adult axonal plasticity and regrowth after CNS injury. J Neurosci. 2006;26(12):3087–3101.[Google Scholar]
  • 70. Ciossek T, et al Eph receptor-ligand interactions are necessary for guidance of retinal ganglion cell axons in vitro. Eur J Neurosci. 1998;10(5):1574–1580.[PubMed][Google Scholar]
  • 71. Fu CT, Sretavan DInvolvement of EphB/Ephrin-B signaling in axonal survival in mouse experimental glaucoma. Invest Ophthalmol Vis Sci. 2012;53(1):76–84.[Google Scholar]
  • 72. Ethell IM, Hagihara K, Miura Y, Irie F, Yamaguchi YSynbindin, A novel syndecan-2-binding protein in neuronal dendritic spines. J Cell Biol. 2000;151(1):53–68.[Google Scholar]
  • 73. Ethell IM, Yamaguchi YCell surface heparan sulfate proteoglycan syndecan-2 induces the maturation of dendritic spines in rat hippocampal neurons. J Cell Biol. 1999;144(3):575–586.[Google Scholar]
  • 74. Ethell IM, et al EphB/syndecan-2 signaling in dendritic spine morphogenesis. Neuron. 2001;31(6):1001–1013.[PubMed][Google Scholar]
  • 75. Zeng Q, et al A novel synaptobrevin/VAMP homologous protein (VAMP5) is increased during in vitro myogenesis and present in the plasma membrane. Mol Biol Cell. 1998;9(9):2423–2437.[Google Scholar]
  • 76. Wang CC, et al A role of VAMP8/endobrevin in regulated exocytosis of pancreatic acinar cells. Dev Cell. 2004;7(3):359–371.[PubMed][Google Scholar]
  • 77. Wang CC, et al VAMP8/endobrevin as a general vesicular SNARE for regulated exocytosis of the exocrine system. Mol Biol Cell. 2007;18(3):1056–1063.[Google Scholar]
  • 78. Ho YH, Cai DT, Huang D, Wang CC, Wong SHCaspases regulate VAMP-8 expression and phagocytosis in dendritic cells. Biochem Biophys Res Commun. 2009;387(2):371–375.[PubMed][Google Scholar]
  • 79. Sherry DM, Wang MM, Frishman LJDifferential distribution of vesicle associated membrane protein isoforms in the mouse retina. Mol Vis. 2003;9:673–688.[PubMed][Google Scholar]
  • 80. Morgans CW, et al Loss of the Synaptic Vesicle Protein SV2B results in reduced neurotransmission and altered synaptic vesicle protein expression in the retina. PLoS ONE. 2009;4(4):e5230.[Google Scholar]
  • 81. De Strooper B, et al A presenilin-1-dependent gamma-secretase-like protease mediates release of Notch intracellular domain. Nature. 1999;398(6727):518–522.[PubMed][Google Scholar]
  • 82. Kopan R, Ilagan MXGamma-secretase: Proteasome of the membrane? Nat Rev Mol Cell Biol. 2004;5(6):499–504.[PubMed][Google Scholar]
  • 83. Sisodia SS, St George-Hyslop PHgamma-Secretase, Notch, Abeta and Alzheimer’s disease: Where do the presenilins fit in? Nat Rev Neurosci. 2002;3(4):281–290.[PubMed][Google Scholar]
  • 84. Wang R, Tang P, Wang P, Boissy RE, Zheng HRegulation of tyrosinase trafficking and processing by presenilins: Partial loss of function by familial Alzheimer’s disease mutation. Proc Natl Acad Sci USA. 2006;103(2):353–358.[Google Scholar]
  • 85. Esselens C, et al Presenilin 1 mediates the turnover of telencephalin in hippocampal neurons via an autophagic degradative pathway. J Cell Biol. 2004;166(7):1041–1054.[Google Scholar]
  • 86. Parent A, Linden DJ, Sisodia SS, Borchelt DRSynaptic transmission and hippocampal long-term potentiation in transgenic mice expressing FAD-linked presenilin 1. Neurobiol Dis. 1999;6(1):56–62.[PubMed][Google Scholar]
  • 87. Barnes S, Kelly MECalcium channels at the photoreceptor synapse. Adv Exp Med Biol. 2002;514:465–476.[PubMed][Google Scholar]
  • 88. Strom TM, et al An L-type calcium-channel gene mutated in incomplete X-linked congenital stationary night blindness. Nat Genet. 1998;19(3):260–263.[PubMed][Google Scholar]
  • 89. Bech-Hansen NT, et al Loss-of-function mutations in a calcium-channel alpha1-subunit gene in Xp11.23 cause incomplete X-linked congenital stationary night blindness. Nat Genet. 1998;19(3):264–267.[PubMed][Google Scholar]
  • 90. Wycisk KA, et al Mutation in the auxiliary calcium-channel subunit CACNA2D4 causes autosomal recessive cone dystrophy. Am J Hum Genet. 2006;79(5):973–977.[Google Scholar]
  • 91. Zhang L, Liu T, Huang Y, Liu JmicroRNA-182 inhibits the proliferation and invasion of human lung adenocarcinoma cells through its effect on human cortical actin-associated protein. Int J Mol Med. 2011;28(3):381–388.[PubMed][Google Scholar]
  • 92. Ebrey T, Koutalos YVertebrate photoreceptors. Prog Retin Eye Res. 2001;20(1):49–94.[PubMed][Google Scholar]
  • 93. Szél A, et al Unique topographic separation of two spectral classes of cones in the mouse retina. J Comp Neurol. 1992;325(3):327–342.[PubMed][Google Scholar]
  • 94. Applebury ML, et al The murine cone photoreceptor: A single cone type expresses both S and M opsins with retinal spatial patterning. Neuron. 2000;27(3):513–523.[PubMed][Google Scholar]
  • 95. Ng L, et al A thyroid hormone receptor that is required for the development of green cone photoreceptors. Nat Genet. 2001;27(1):94–98.[PubMed][Google Scholar]
  • 96. Fei YDevelopment of the cone photoreceptor mosaic in the mouse retina revealed by fluorescent cones in transgenic mice. Mol Vis. 2003;9:31–42.[PubMed][Google Scholar]
  • 97. Fujieda H, Bremner R, Mears AJ, Sasaki HRetinoic acid receptor-related orphan receptor alpha regulates a subset of cone genes during mouse retinal development. J Neurochem. 2009;108(1):91–101.[PubMed][Google Scholar]
  • 98. Swaroop A, et al A conserved retina-specific gene encodes a basic motif/leucine zipper domain. Proc Natl Acad Sci USA. 1992;89(1):266–270.[Google Scholar]
  • 99. Mitton KP, et al. The leucine zipper of NRL interacts with the CRX homeodomain. A possible mechanism of transcriptional synergy in rhodopsin regulation. J Biol Chem. 2000;275(38):29794–29799.[PubMed]
  • 100. Pittler SJ, et al Functional analysis of the rod photoreceptor cGMP phosphodiesterase alpha-subunit gene promoter: Nrl and Crx are required for full transcriptional activity. J Biol Chem. 2004;279(19):19800–19807.[PubMed][Google Scholar]
  • 101. Yoshida S, et al Expression profiling of the developing and mature Nrl-/- mouse retina: Identification of retinal disease candidates and transcriptional regulatory targets of Nrl. Hum Mol Genet. 2004;13(14):1487–1503.[PubMed][Google Scholar]
  • 102. Oh EC, et al Rod differentiation factor NRL activates the expression of nuclear receptor NR2E3 to suppress the development of cone photoreceptors. Brain Res. 2008;1236:16–29.[Google Scholar]
  • 103. Hennig AK, Peng GH, Chen SRegulation of photoreceptor gene expression by Crx-associated transcription factor network. Brain Res. 2008;1192:114–133.[Google Scholar]
  • 104. Baylor DA, Lamb TD, Yau KWThe membrane current of single rod outer segments. J Physiol. 1979;288:589–611.[Google Scholar]
  • 105. Rich KA, Zhan Y, Blanks JCMigration and synaptogenesis of cone photoreceptors in the developing mouse retina. J Comp Neurol. 1997;388(1):47–63.[PubMed][Google Scholar]
  • 106. Miyake Y, Yagasaki K, Horiguchi M, Kawase Y, Kanda T. Congenital stationary night blindness with negative electroretinogram. A new classification. Arch Ophthalmol. 1986;104(7):1013–1020.[PubMed]
  • 107. Yan D, Liu XZGenetics and pathological mechanisms of Usher syndrome. J Hum Genet. 2010;55(6):327–335.[Google Scholar]
  • 108. Xu S, et al PHR1, a PH domain-containing protein expressed in primary sensory neurons. Mol Cell Biol. 2004;24(20):9137–9151.[Google Scholar]
  • 109. Nagy A, Gertsenstein M, Vintersten K, Behringer R Manipulating the Mouse Embryo. A Laboratory Manual. 3rd Ed. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory; 2003. [PubMed][Google Scholar]
  • 110. Perez SE, Lumayag S, Kovacs B, Mufson EJ, Xu SBeta-amyloid deposition and functional impairment in the retina of the APPswe/PS1DeltaE9 transgenic mouse model of Alzheimer’s disease. Invest Ophthalmol Vis Sci. 2009;50(2):793–800.[Google Scholar]
  • 111. Peachey NS, Goto Y, al-Ubaidi MR, Naash MIProperties of the mouse cone-mediated electroretinogram during light adaptation. Neurosci Lett. 1993;162(1-2):9–11.[PubMed][Google Scholar]
  • 112. Xu S, et al The proliferation and expansion of retinal stem cells require functional Pax6. Dev Biol. 2007;304(2):713–721.[Google Scholar]
  • 113. Ye X, et al Norrin, frizzled-4, and Lrp5 signaling in endothelial cells controls a genetic program for retinal vascularization. Cell. 2009;139(2):285–298.[Google Scholar]
  • 114. Nicholson DA, Geinisman YAxospinous synaptic subtype-specific differences in structure, size, ionotropic receptor expression, and connectivity in apical dendritic regions of rat hippocampal CA1 pyramidal neurons. J Comp Neurol. 2009;512(3):399–418.[Google Scholar]
  • 115. Young RWCell death during differentiation of the retina in the mouse. J Comp Neurol. 1984;229(3):362–373.[PubMed][Google Scholar]
  • 116. Xu S, Witmer PD, Lumayag S, Kovacs B, Valle DMicroRNA (miRNA) transcriptome of mouse retina and identification of a sensory organ-specific miRNA cluster. J Biol Chem. 2007;282(34):25053–25066.[PubMed][Google Scholar]
  • 117. Zhu Q, et al Sponge transgenic mouse model reveals important roles for the microRNA-183 (miR-183)/96/182 cluster in postmitotic photoreceptors of the retina. J Biol Chem. 2011;286(36):31749–31760.[Google Scholar]
  • 118. McCall MA, Gregg RGComparisons of structural and functional abnormalities in mouse b-wave mutants. J Physiol. 2008;586(Pt 18):4385–4392.[Google Scholar]
  • 119. Rich KA, Zhan Y, Blanks JCMigration and synaptogenesis of cone photoreceptors in the developing mouse retina. J Comp Neurol. 1997;388(1):47–63.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.