Molecular Vision 2020; 26:173-187 <http://www.molvis.org/molvis/v26/173>
Received 11 October 2019 | Accepted 04 March 2020 | Published 06 March 2020

Using BXD mouse strains in vision research: A systems genetics approach

Eldon E. Geisert,1 Robert W. Williams2

1Department of Ophthalmology, Emory University, 1365B Clifton Road NE Atlanta GA, 30322; 2Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, 71 S Manassas St, Memphis TN 38163

Correspondence t o: Eldon E. Geisert, Department of Ophthalmology, Emory University, Room B5512, 1365B Clifton Road N.E., Atlanta, GA, 30322; Phone: (404) 778 4239; FAX: (404) 778-2231; email: egeiser@emory.edu

Abstract

We illustrate the growing power of the BXD family of mice (recombinant inbred strains from a cross of C57BL/6J and DBA/2J mice) and companion bioinformatic tools to study complex genome-phenome relations related to glaucoma. Over the past 16 years, our group has integrated powerful murine resources and web-accessible tools to identify networks modulating visual system traits—from photoreceptors to the visual cortex. Recent studies focused on retinal ganglion cells and glaucoma risk factors, including intraocular pressure (IOP), central corneal thickness (CCT), and susceptibility of cellular stress. The BXD family was exploited to define key gene variants and then establish linkage to glaucoma in human cohorts. The power of this experimental approach to precision medicine is highlighted by recent studies that defined cadherin 11 (Cdh11) and a calcium channel (Cacna2d1) as genes modulating IOP, Pou6f2 as a genetic link between CCT and retinal ganglion cell (RGC) death, and Aldh7a1 as a gene that modulates the susceptibility of RGCs to death after elevated IOP. The role of three of these gene variants in glaucoma is discussed, along with the pathways activated in the disease process.

Introduction

Since the first wave of whole genome sequencing in the early 2000s [1-7], teams of investigators have developed open genetic, genomic, and transcriptomic resources to study the eye, visual system, and blinding diseases [8-12]. Over this same period, the costs of generating high-quality phenome and genome data have been decreased, and data quality and throughput have improved greatly. We are now at the point that is practical to consider in-depth analyses across large cohorts of human populations and rodent models. In the case of isogenic cohorts of mice and rats, the analysis of single genomes can be extended to multiple time points during development, aging, and in the progression of blinding diseases, making it possible to study gene-by-environmental and gene-by-treatment effects in ways that have high translational relevance to human clinical disease. A second major advance is our ability to systematically define and validate causal gene variants with increasing precision, power, and efficiency [13,14]. We can then move on to the more important stage of research to probe linked molecular and cellular networks associated with variation in the eye and visual system structure, function, disease, and treatment. For the first time, we can, in principle, combine a systems approach across the entire visual system (from the cornea to the cortex to visually guided behavior) using global omics and genetic methods, including epigenomic, proteomic, metabolomic, and lipidomic methods. When this omics combination is coupled with classic genetics (i.e., genetically diverse populations), it is referred to as systems genetics; essentially systems biology but in a rich genetics and omics context [2,3,15].

Progress in systems genetics was driven by the development of greatly expanded families of fully isogenic replicable cohorts [16,17], in particular the BXD strain set (parental strains, the C57BL/6J mouse and the DBA/2J mouse, Figure 1) [18,19], the AXB/BXA strain set (parental strains, the AJ mouse and the C57BL/6J mouse) [20], and the collaborative cross (CC,) [21,22]. In addition, the CC is used to create the mouse diversity outbred strains [23,24]. These recombinant inbred strains have been used in studies of the visual system, focusing mainly on the retina [17,25-28]. Table 1 lists the attributes of each strain and the strain set–specific tools available for data analysis. Each recombinant inbred strain has its advantages. The AXB strains were used extensively to map quantitative trait loci (QTLs) modulating cell number in the retina [29,30]. The disadvantage of this strain set is that there are a limited number of strains (now in cryopreservation) and only one data set for transcriptome analysis of the whole eye. The CC has the advantage of an extremely diverse genetic background with eight parental mouse strains contributing to the overall genetic diversity. Seventy-five strains are available from the Jackson Laboratory; however, this strain set is not widely used in vision research. All of the recombinant inbred strains have the distinct advantage of resampling the same genetic background. This ability to resample decreases the contribution of environmental variance and dramatically enhances the ability to accurately map QTLs. The fact that mice cannot be resampled in the mouse diversity panel is a decidedly distinct disadvantage.

At the present time, the BXD family is one of the preeminent platforms for systems biology to study the visual system and neurodegeneration [12,15,27,31-34]. The BXD family consists of 150 strains that segregate at more than 6 million sequence variants—a level comparable to many human cohorts used in genome-wide association studies (GWASs) [19]. There are several advantages vis-à-vis studies of human cohorts: efficient experimental procedures and therapeutics, access to cells and tissues at any time point under many controlled conditions, high statistical power (resampling individual isogenic genotypes), high mapping precision (often better than 2 mega-bases), the ability to study gene-by-environmental interactions, and faster exploration and testing of the potential and the limitations of precision medicine.

The BXD family (Figure 1) was derived by crossing two of the most widely used inbred strains of mice: C57BL/6J (B6) and DBA/2J (D2). The B6-by-D2 offspring (BXD) were then inbred along separate lines until each line was fully inbred. Each of the 150 BXD progeny strains is essentially immortal and a fully inbred sibling. Some of these strains have been used for nearly 25 years for rigorous quantitative analysis of the visual system and the retina structure [35], cortical plasticity [27,36], as well as for studies of eye and retinal transcriptomes [1,37].

The development of the BXD family was begun by Benjamin A. Taylor in about 1973. He generated the two sets of these strains (BXD1 to BXD32 [38], and then BXD33 to BXD42 [18]) at the Jackson Laboratory. BXD43 through BXD102 were generated by Lu Lu, Jeremy Pierce, and colleagues in the late 1990s and early 2000s using advanced intercross progeny [39]. Recent efforts by our group have increased the number of BXD progeny to 150 living strains. All are currently available from either the Jackson Laboratory or the University of Tennessee Health Science Center. All have been fully sequenced (this is publicly available) [19].

The BXD family incorporates a comparatively high level of genetic diversity and can serve as a robust animal model for some human ophthalmic diseases and developmental abnormalities. The use of sophisticated molecular, imaging, and phenotyping methods across such a large set of fully sequenced and isogenic (reproducible) lines of mice opens up new opportunities in an experimental version of precision medicine. We focus on the eye, retina, and primary visual system. The BXD family of strains offers a unique resource for the vision research community with several advantages. Across the strains, there is a relatively high level of diversity in phenotypes. For example, the total population of retinal ganglion cells per eye varies from about 50,800 ± 1,100 in BXD27 to 75,800 ± 2,000 in BXD32 [40]. The practicality of resampling each genome many times—for example, to gain precise estimates of RGC numbers—greatly reduces non-genetic sources of variance and boosts the effective heritability of traits. This makes it possible to map and even clone the most stubborn and noisiest phenotypes. Initial studies by our group explored the genetic diversity within the BXD family to define genetic, molecular, and phenotypic networks active in the eye (Table 2).

We have developed companion open access data resources and analytic tools that make the statistical and mapping methods far more accessible to a large community of vision research scientists who have matched expertise in molecular and cellular biology (for detailed instruction on the use of these resources, see Geisert et al. [1]). For example, we generated, collected, and curated multiple gene expression data sets (Table 2). Normative and experimental data sets facilitate the study of ocular diseases and injury in eyes [1] and the retinas [11,27,37,41]. The complete eye data set is the Eye M430v2 (Sep08) RMA which contains data from 68 BXD RI strains, the parental strain, the reciprocal F1 crosses, 35 strains from the mouse diversity panel, and eye data from six knockout mouse lines. The other data sets are subsets taken from the original data. One of the parental strains contains mutations in two genes (Tyrp1 and Gpnmb) that contribute to pigment dispersion and ultimately, to glaucoma [42-44]. The derivative data sets are split subsamples based on the presence or absence of mutations in these two genes. For example, the Eye M430v2 no Mutant/Mutant (Aug12) RMA data set contains array data for eyes from 57 BXD strains, none of which have mutations in Tyrp1 or Gpnmb.

There are also several different retinal transcriptome data sets. The most recent exploits the Affymetrix Mouse Gene 2.0 ST Exon array (Santa Clara, CA) to estimate change in gene expression between the healthy retina and the retina 5 days after the eye was exposed to a 50 psi blast injury ((Table 2) [37,41]. These Affymetrix data sets include the DoD Retina Normal Affy MoGene 2.0 ST (May15) RMA Gene Level (the data set was made using healthy retinas from 59 different mouse strains), DoD Retina Normal Affy MoGene 2.0 ST (May15) RMA Exon Level (the data set explores 59 strains at the exon level), DoD Retina After Blast Affy MoGene 2.0 ST (March16) RMA Gene Level (the data set consists of gene-level data from the retinas of 54 strains 5 days following blast injury), DoD Retina After Blast Affy MoGene 2.0 ST (March16) RMA Exon Level (the 5-day blast injury data are also presented at the exon level), and DoD Retina Blast versus Normal Affy MoGene 2.0 ST (April15) RMA Gene Level. The other data sets were run on the Illumina V6.2 array (San Diego, CA), and the difference in gene expression between healthy retinas and retinas 2 days after optic nerve crush was examined [11,27]. The arrays in the Illumina data set include Full HEI Retina Illumina V6.2 (April10) RankInv (the data set represents gene-level expression data from the healthy retinas from 75 BXD strains), HEI Retina Normal Illumina V6.2 (April10) RankInv (the data set is similar to that for the full retina with the removal of data from six strains with high levels of glial fibrillary acidic protein (GFAP) expression), and ONC HEI Retina (April12) RankInv (the data set consists of retinal samples 2 days after optic nerve crush). These BXD data sets provide a large resource that is especially useful for characterizing molecular and genetic networks in the eye, and for tracking down sequence variants related to the injury or disease susceptibility [1,11,45,46]. Furthermore, all the retinal mRNA profile data from healthy and injured conditions are accessible by the public on GeneNetwork, along with a sophisticated array of bioinformatic tools.

Defining genetic networks active in the eye and the retina

The eye and retina transcriptome data sets provide powerful tools for defining the genetic profiles of specific ocular tissues and cell types [1,47,48]. In the HEI Eye Data Set, signature transcript profiles can define the genes expressed in the cornea to retinal ganglion cells [1]. The large microarray data sets also allow us to define genetic networks active within the retina and the changes in these networks that occur after injury [11,49]. One example is the activation of an innate immune network within the retina following optic nerve crush or blast injury to the eye. Using four comprehensive and complementary transcriptome data sets (the healthy retina and the retina 2 days after optic nerve crush, along with the healthy retina and the retina 5 days after blast injury, gn2.GeneNetwork.org), we examined changes that occur in gene expression profiles after optic nerve crush or after blast injury. Our group found an innate immune network that is rapidly activated after injury to the retina [11,41]. This work added to the previous work of others that showed members of the complement cascade are involved in retinal injury and glaucoma [50-53]. The importance of this complement network in glaucoma was revealed by knocking out C1qa on a DBA/2J background. In these animals, pigmentary dispersion glaucoma and elevated intraocular pressure (IOP) occur, but the expected loss of axons in the optic nerve is dramatically mitigated [53]. Thus, C1QA, and potentially, the complement cascade, plays a pivotal role in the degeneration of axons within the optic nerve of the DBA/2J glaucoma mouse model.

An examination of the changes in the expression of C1qa and C4 following optic nerve crush or blast injury led to the identification of a genetic network activated by injury. The prominent genes in this network are members of the innate immune system. Defining the activation of these genetic pathways was made possible by the large number of BXD strains in these data sets [1,11]. The power of this research effort allows us to define genetic networks in the healthy mouse retina and genetic networks activated by injury (based on 61 BXD strains). If we look at the distribution of C4b across the BXD RI strain set, we can see that selected animals have high levels of expression. If we compare the top 100 correlates of C4b in the injured retina to those in the healthy retina, then there is clear upregulation of the expression of these genes, and the genes are more highly correlated than observed in the healthy retina database (Figure 2). Examining cellular markers in the activated network, it appears that retinal microglia are the main cellular component of the innate immune response in the retina [11]. Defining the innate immune genetic network in the retina illustrates the power of the BXD strain set to examine the coordinated activation of genomic elements.

Systems biology of ocular phenotypes

One of the primary uses of the BXD family is to map QTLs that modulate ocular phenotypes or functional aspects of the visual system. Our group has conducted studies on several morphometric features, beginning with ganglion cell number [35], eye size, lens weight, and retinal area [54]. Examination of one of the targets of the retina, the lateral geniculate nucleus (LGN), in the BXD strains, led to interesting correlations between the number of RGC neurons and the number of neurons in the lateral geniculate nucleus. The study of the LGN is a good example of how systematically generating quantitative data across the BXD family can lead to substantial revisions in our understanding of the visual system [55]. Before the LGN work, it was universally thought that the numbers of RGC neurons and neurons in the LGN were jointly titrated during development to some optimal functional stoichiometry—for example, to a relative tightly adjusted ratio between RGC neurons and their principal targets in the LGN. However, Seecharan and colleagues [55] refuted this neat numerical matching hypothesis. Across 56 strains, the correlation between the numbers of RGC and LGN neurons is merely 0.01, strong evidence that interconnected neurons in these two regions are not jointly controlled by either genetic or developmental mechanisms. Other families of inbred strains were used to examine other cell types within the retina [30,48,56-61]. Finally, recent studies used the BXD strain set to examine the regenerative capacity of axons in the optic nerve [62]. All of the phenotypic data are available on GeneNetwork under BXD Phenotypes.

The tools and databases presented in GeneNetwork were used to examine genetic regulation of factors associated with glaucoma risk in humans [63]. Glaucoma affects millions of people worldwide [64,65] and is the second leading cause of blindness in the United States [66]. Adult-onset glaucoma is a collection of diseases with multiple risk factors and genes that ultimately affect the loss of RGCs [67-69]. The severity of primary open angle glaucoma (POAG) is dependent on the interaction of multiple genes, age, and environmental factors [70]. The primary risk factor is elevated IOP [71]. There are known genetic mutations that affect IOP that result in inherited glaucoma [72,73]. Since the Ocular Hypertension Treatment Studies (OHTS) [63] and others’ subsequent independent findings [74,75], several phenotypic risk factors for POAG have been identified. Two of these glaucoma risk factors are IOP and central corneal thickness (CCT). We identified genes modulating these phenotypic factors in the mouse and examined human glaucoma to determine the potential role of these genes in human populations. As an alternative approach to human GWASs, we used mouse model systems to define genes regulating ocular phenotypes and potential links between these ocular traits and glaucoma risk [76]. These mouse models aid not only in defining genes involved in glaucoma risk but also in understanding the disease mechanism along with potential therapeutic interventions [77,78].

Regulation of IOP

The BXD family was used in three studies to examine QTLs modulating IOP in the mouse [12,25,28]. We know a considerable amount about the regulation of IOP from the production of aqueous humor to the outflow pathways [79,80]. We also know that IOP is a complex trait affected by different tissues in the eye each of which may be regulated by multiple genes. Interestingly, until recently few studies [81-86] had identified genomic loci in humans modulating healthy IOP. Recent work using the eye phenotypic data from the UK Biobank identified several genes modulating IOP in human populations [87-89]. Lu et al. [28] examined the regulation of IOP in BXD strains relative to pigment dispersion (transillumination deficit), time of day, and age. The study found that IOP across the full array of BXD strains was independent of transillumination defects or the time of day. There was no single genomic locus identified in this study that was found to modulate IOP in this set of BXD strains. The second study by Chintalapudi et al. [25] examined 65 BXD strains and found a significant QTL peak on chromosome 5. Within this QTL, one gene (Cacna2D1) was identified as a likely candidate for modulating IOP in the mouse. When the , National Eye Institute Glaucoma Human genetics collaBORation Heritable Overall Operational Database (NEIGHBORHOOD) GWAS was examined, calcium channel, voltage-dependent, alpha 2/delta subunit 1 (CACNA2D1-HGNC: 1399 Entrez Gene: 781 Ensembl: ENSG00000153956 OMIM: 114204) had a nominal association with POAG (p<0.001). This protein was also found to be highly expressed in the ciliary body and displayed expression in the trabecular meshwork. The identification of this candidate gene, Cacna2d1, provided a promising new target for therapeutic intervention to modulate IOP in POAG. The third study by King et al. [12] used 38 BXD strains, none of which carried the two mutations (Tyrp1 and Gpnmb) that result in pigment dispersion and elevated IOP. The genome-wide interval map identified one significant peak on chromosome 8 that lies in a gene desert. Within this region, there are only four annotated genes: Gm15679 (predicted gene 15,679), Cdh8 (cadherin 8), Cdh11 (cadherin 11), and Gm8730 (predicted gene 8730). Defining good candidate genes within the QTL requires a genomic element that eventually affects protein function. There are several possibilities: an increase or decrease in the amount of transcript produced by each strain, resulting in differences of protein expression or a difference in RNA levels (specifically non-coding RNAs or microRNAs). Another possibility is there is a mutation in the transcript that affects its function. For a protein, this would represent a non-synonymous single nucleotide polymorphism (SNP) that has a deleterious effect on protein function. For RNA, it would represent a sequence change that alters the function of the RNA. Using the tools available on GeneNetwork, we were able to identify candidate genes, particularly those associated with so-called cis-QTLs [1] or with nonsynonymous SNPs changing protein sequence and affecting protein function [47]. There are only two strong candidate genes, Cdh11 and Cdh8. Neither has cis-QTLs in eye or retinal data sets [1,37]. Both have non-synonymous SNPs. Based on expression levels and on Sorting Intolerant from Tolerant (SIFT) analysis (a test designed to define SNPs that affect protein function), Cdh11 was the single strongest candidate. Examining human RNA sequencing (RNA-seq) data for the trabecular meshwork [90], we found expression of CDH11 is 55 times higher than that of CDH8. Cadherin 11 was also found in structures associated with the control of IOP. In sections of the mouse eye stained for cadherin 11, there was antibody-specific staining of the trabecular meshwork, the endothelial cells of the canal of Schlemm, and other structures within the angle of the eye. The expression pattern of cadherin 11 in the cells of the trabecular meshwork and the canal of Schlemm is appropriate for a protein involved in regulation of IOP. After we found these results in the mouse, two human GWASs identified cadherin 11 (CDH11-HGNC: 1750 Entrez Gene: 1009 Ensembl: ENSG00000140937 OMIM: 600023) as a gene involved in the regulation of IOP in humans [87,88]. Recent studies demonstrated that CDH11 is also a glaucoma risk factor [91]. These data revealed that cadherin 11 is a modulator of IOP and a risk for glaucoma.

Cadherins play an important role in cell–cell adhesion [92-94], and trabecular meshwork cells express several family members, including VE-cadherin (cadherin 5), K-cadherin (cadherin 6), OB-cadherin (cadherin 11), cadherin 19, and N-cadherin (cadherin 2) [95-97]. In the trabecular meshwork, cadherins can be modulated by TGFβ [98]. In culture, treating trabecular meshwork cells with Wnt3a causes an increase in cadherin 11 expression, resulting in enhanced cell–cell adhesion. The levels of cadherin 11 can also be affected by the wingless (Wnt)/β-catenin pathway [99]. Wnt signaling is known to increase membrane associated cadherin 6 [100]. The TGFβ and Wnt/β-catenin pathways interact with each other in modulating cadherin expression, specifically cadherin 2, cadherin 6, and cadherin 11. The upregulation of these cadherins in cultured trabecular meshwork cells enhances the adhesion between the cells resulting in an increase in resistance across the culture cell monolayers. The interaction between the Wnt signaling pathway and cadherins appears to be important in the regulation of IOP. The Clark group [101] found that increasing TGFβ elevates IOP while the Wnt pathway maintains IOP homeostasis. We have shown that Cdh11 modulates IOP in the mouse [34]. Others have implicated CDH11 in IOP regulation in humans [87,88]. It appears to be part of an integral network involved in TGFβ and the Wnt/β-catenin signaling that modulates cell adhesion between trabecular meshwork cells. These interactions play an important role in the regulation of IOP and potentially form the basis of the role of CDH11 in glaucoma [91].

Susceptibility of RGCs to injury

To define genomic elements modulating the susceptibility of RGCs to injury, we examined axon loss in 49 BXD strains that had magnetic beads injected into the anterior chamber of one eye blocking the trabecular meshwork and elevating IOP [49]. When the number of axons in the healthy retina and the number of axons following elevated IOP were used to generate genome-wide interval maps, they revealed the same suggestive QTLs on proximal chromosome 3. Neither the healthy nerve nor the optic nerve after elevated IOP had QTLs that reached a level of statistical significance (p>0.05). To define genomic loci that could modulate the susceptibility of RGCs to death, the loss of axons per strain was calculated by subtracting the mean number of axons in bead-injected eyes from the mean number of axons in healthy eyes for each strain. The genome-wide interval map revealed a single statistically significant genomic locus on chromosome 18 (54 to 56 Mb). Within this locus, there were no non-synonymous SNPs that could account for the allelic differences between the C57BL/6J and DBA/2J mice. One gene, Aldh7a1, had a significant linkage related score (LRS = 31, p<0.01, Probe 17,354,434). Aldh7a1 was the single cis-eQTL within this interval.

The distribution of the ALDH7A1 protein in retinal sections or flatmounts reveals it is within RGCs that are colabeled with RGC markers TUJ1 or RBPMS [48,102]. ALDH7A1 staining was relatively ubiquitous in the cell body and axons but absent from the nucleus. These results are in line with other studies that demonstrated mitochondrial and cytosolic localization of ALDH7A1 in humans and rodents [103-105].

It is interesting to speculate about the potential role of ALDH7A1 in glaucoma risk. ALDH7A1 is involved in the metabolism of acetaldehyde to acetic acid. This process also involves metabolism of NAD. This coregulation of NAD may have a direct effect on axon and neuronal survival. These data suggest that allelic differences in ALDH7A1 may affect mitochondria function resulting in the susceptibility of RGCs to death. Previous work revealed that mitochondrial function is critical for RGC survival. The prime example is an interesting murine mutation that affects Wallerian degeneration in the peripheral nervous system [106] and the central nervous system [107]. The mutation disrupting healthy axon degeneration is a chimeric protein made up of Ube4b and Nmnat1 that produces the Wallerian Degeneration Slow (Wlds) protein. The effect of Wlds on axonal degeneration is due to local activity in the axon itself. The chimeric protein localizes not only to the nucleus but also in small axonal pools [108], causing a local increase in NAD+. In a rat model of glaucoma, Wlds was shown to protect axons from degeneration, but did not appear to alter the fate of the neuronal cell bodies [109]. In the D2 mouse model of glaucoma, supplementing the diet with NAD or overexpressing Nmnat1 partially protects against glaucomatous degeneration [110]. When the mutation in WLDs was put on the D2 background, and the mouse was supplemented with NAM (nicotinamide, a NAD precursor), there was almost complete rescue from the effects of glaucoma with 94% of the treated eyes not developing glaucoma [78]. It is possible that the interactions of ALDH7A1 with NAD are in part responsible for the glaucoma risk inferred by specific mutations in this protein.

Central corneal thickness and glaucoma

The BXD strains were used to define a genetic link between CCT and glaucoma risk [12]. CCT is one of the most heritable ocular phenotypes, and it is also a risk for developing POAG [111-113]. Thinner corneas are associated with an increased risk of developing POAG, and this risk is independent of the confounding effects of CCT on intraocular pressure measurements [63,75]. A thinner CCT is also associated with increased severity of visual field loss and more rapid progression of the disease [66,70,114]. Pou6f2 was identified as a gene that modulates CCT in the mouse, and in the NEIGHBORHOOD human glaucoma database, POU6F2 (HGNC: 21694 Entrez Gene: 11281 Ensembl: ENSG00000106536 OMIM: 609062) is a risk factor for human glaucoma [91,115,116].

CCT was measured in 61 BXD RI strains (Figure 3), and a single significant QTL (Figure 3) was identified on chromosome 13 (13 to 19 Mb). Within this QTL, there was only one candidate gene Pou6f2 that contained non-synonymous SNPs in the mouse. The syntenic regions in the human were examined by Wiggs and colleagues in the NEIGHBORHOOD database [117] to determine if there are potential risk factors for glaucoma in this region. The top 50 SNPs were all associated with one gene (POU6F2). The highest statistically significant level was a probability of 10−6 for SNP rs76319873. This combined approach identified Pou6f2 as a gene that modulates CCT in the mouse and a risk factor for primary open angle glaucoma [91].

POU6F2 was first described as a novel POU-domain transcription factor in the retina [118], and it identified a subpopulation of RGCs. We have independently confirmed these findings and found that Pou6f2 is part of a genetic network found in mouse RGCs [48]. The first hint of the link between Pou6f2 modulating CCT in the mouse and the potential role of the gene in glaucoma is revealed during the development of the eye. POU6F2 is expressed in RGC progenitor cells and the cornea. In the embryonic eye, strong POU6F2 staining was observed in neuroblasts destined to become RGCs. There is also staining of the developing cornea and corneal stem cells [12].

In flatmounts of the mouse retina, virtually all of the POU6F2-positive cells are labeled with RNA-binding protein with multiple splicing, RBPMS (Figure 4). There are cells that are heavily labeled (approximately 16% of RPBMS RGCs) and cells that are moderately to lightly labeled (approximately 16% of the RPBMS RGCs). A few cells in the amacrine cell layer are POU6F2 positive, and all of these cells are also labeled with RBPMS, suggesting that these cells are displaced ganglion cells. In retinas 28 days following optic nerve crush, no cells in the ganglion cell layer are positive for POU6F2. As POU6F2 is found only in RBPMS positive cells, and as all of the staining for POU6F2 disappears following optic nerve crush, we conclude that POU6F2 labels only RGCs in the C57BL/6J and DBA2J mouse adult retinas.

To examine the potential role of POU6F2 in glaucoma, we compared the distribution of POU6F2-RGCs in four young D2 mice (70 days old) to four older D2 mice (8 months old). There was a 22% loss of RPBMS labeled RGCs in the aged mice, while there was a 73% loss of POU6F2 heavily labeled cells and a 10% loss of POU6F2 moderate to lightly labeled RGCs (Figure 4). These data demonstrate that heavily labeled POU6F2 RGCs are sensitive to early phases of glaucoma in the DBA/2J mouse model.

Conclusions

The BXD family and the research tools developed on GeneNetwork offer the vision research community a unique system for analyzing complex genomic interactions associated with the healthy development of the mammalian visual system and disease. The initial efforts of our group explored the genetic diversity within the BXD RI strain set and defined genetic networks active in the eye. As a result of this work, we provided the vision research community with the Hamilton Eye Institute Mouse Eye Database (HEIMED [1]). To continue our efforts in studying the complex biology and diseases of the eye, we created the HEI Retinal Database. Within this database, naturally occurring changes in the mRNA levels are defined as the phenotype, and the genomic loci modulating the differences in transcriptional control can be evaluated using traditional QTL mapping methods. Thus, the HEI Retinal Database and the DoD Normal Retina Database provide a transcriptome-wide analysis of the retina, which allows identification of the genetic variability between the BXD RI strains and the expression signatures of cells that underlie the phenotypic variation. The utility of the BXD strains and the expression databases offered on GeneNetwork is demonstrated by the identification of Pou6f2 as a modulator of CCT (a risk factor for POAG) and Cdh11 as a modulator of IOP (a risk factor for POAG). Both genes were recently identified as glaucoma risk factors [91] providing a genetic link between CCT and IOP to glaucoma risk.

Acknowledgements:

This study was supported by an Unrestricted Grant from Research to Prevent Blindness, NEI grant R01EY017841 (E.E.G.), Owens Family Glaucoma Research Fund, P30EY06360 (Emory Vision Core) and DoD CDMRP Grant W81XWH-12-1-0255 from the USA Army Medical Research & Materiel Command and the Telemedicine and Advanced Technology (E.E.G.). GeneNetwork supported by NIGMS (R01GM123489), NIDA (P30DA044223) and the UT Center for Integrative and Translational Genomics, and funds from the UT-ORNL Governor’s Chair (R.W.W.).

References:

  1. Geisert EE, Lu L, Freeman-Anderson NE, Templeton JP, Nassr M, Wang X, Gu W, Jiao Y, Williams RW. Gene expression in the mouse eye: an online resource for genetics using 103 strains of mice. Mol Vis. 2009; 15:1730-63. [PMID: 19727342]
  2. Mulligan MK, Mozhui K, Prins P, Williams RW. GeneNetwork: A Toolbox for Systems Genetics. Methods Mol Biol. 2017; 1488:75-120. [PMID: 27933521]
  3. Williams RW, Williams EG. Resources for Systems Genetics. Methods Mol Biol. 2017; 1488:3-29. [PMID: 27933518]
  4. Bubier JA, Langston MA, Baker EJ, Chesler EJ. Integrative Functional Genomics for Systems Genetics in GeneWeaver.org. Methods Mol Biol. 2017; 1488:131-52. [PMID: 27933523]
  5. Mardis ER. The impact of next-generation sequencing technology on genetics. Trends Genet. 2008; 24:133-41. [PMID: 18262675]
  6. Metzker ML. Sequencing technologies - the next generation. Nat Rev Genet. 2010; 11:31-46. [PMID: 19997069]
  7. Buermans HP, den Dunnen JT. Next generation sequencing technology: Advances and applications. Biochim Biophys Acta. 2014; 1842:1932-41. [PMID: 24995601]
  8. Roesch K, Stadler MB, Cepko CL. Gene expression changes within Muller glial cells in retinitis pigmentosa. Mol Vis. 2012; 18:1197-214. [PMID: 22665967]
  9. Corbo JC, Myers CA, Lawrence KA, Jadhav AP, Cepko CL. A typology of photoreceptor gene expression patterns in the mouse. Proc Natl Acad Sci USA. 2007; 104:12069-74. [PMID: 17620597]
  10. Blackshaw S, Fraioli RE, Furukawa T, Cepko CL. Comprehensive analysis of photoreceptor gene expression and the identification of candidate retinal disease genes. Cell. 2001; 107:579-89. [PMID: 11733058]
  11. Templeton JP, Freeman NE, Nickerson JM, Jablonski MM, Rex TS, Williams RW, Geisert EE. Innate immune network in the retina activated by optic nerve crush. Invest Ophthalmol Vis Sci. 2013; 54:2599-606. [PMID: 23493296]
  12. King R, Struebing FL, Li Y, Wang J, Koch AA, Cooke Bailey JN, Gharahkhani P. International Glaucoma Genetics C, Consortium N, MacGregor S, Allingham RR, Hauser MA, Wiggs JL, Geisert EE. Genomic locus modulating corneal thickness in the mouse identifies POU6F2 as a potential risk of developing glaucoma. PLoS Genet. 2018; 14:e1007145 [PMID: 29370175]
  13. Putman AH, Wolen AR, Harenza JL, Yordanova RK, Webb BT, Chesler EJ, Miles MF. Identification of quantitative trait loci and candidate genes for an anxiolytic-like response to ethanol in BXD recombinant inbred strains. Genes Brain Behav. 2016; 15:367-81. [PMID: 26948279]
  14. Li H, Wang X, Rukina D, Huang Q, Lin T, Sorrentino V, Zhang H, Bou Sleiman M, Arends D, McDaid A, Luan P, Ziari N, Velazquez-Villegas LA, Gariani K, Kutalik Z, Schoonjans K, Radcliffe RA, Prins P, Morgenthaler S, Williams RW, Auwerx J. An Integrated Systems Genetics and Omics Toolkit to Probe Gene Function. Cell Syst. 2018; 6:90-102. [PMID: 29199021]
  15. Quirós PM, Prado MA, Zamboni N, D’Amico D, Williams RW, Finley D, Gygi SP, Auwerx J. Multi-omics analysis identifies ATF4 as a key regulator of the mitochondrial stress response in mammals. J Cell Biol. 2017; 216:2027-45. [PMID: 28566324]
  16. Sankaran M, Keeley PW, He L, Iuvone PM, Reese BE. Dopaminergic amacrine cell number, plexus density, and dopamine content in the mouse retina: Strain differences and effects of Bax gene disruption. Exp Eye Res. 2018; 177:208-12. [PMID: 30240584]
  17. Keeley PW, Whitney IE, Reese BE. Genomic Control of Retinal Cell Number: Challenges, Protocol, and Results. Methods Mol Biol. 2017; 1488:365-90. [PMID: 27933534]
  18. Taylor BA, Wnek C, Kotlus BS, Roemer N, MacTaggart T, Phillips SJ. Genotyping new BXD recombinant inbred mouse strains and comparison of BXD and consensus maps. Mamm Genome. 1999; 10:335-48. [PMID: 10087289]
  19. Ashbrook DGAD, Prins P, Mulligan MK, Roy S, Williams EG, Lutz CM, Valenzuela A, Bohl CJ. ngels JF, McCarty MS, Centeno AG, Hager R, Auwerx J, Sen S, Lu L, Williams RW. The expanded BXD family of mice: A cohort for experimental systems genetics and precision medicine. bioRxiv. 2019;
  20. Marshall JD, Mu JL, Cheah YC, Nesbitt MN, Frankel WN, Paigen B. The AXB and BXA set of recombinant inbred mouse strains. Mamm Genome. 1992; 3:669-80. [PMID: 1477475]
  21. Churchill GA, Airey DC, Allayee H, Angel JM, Attie AD, Beatty J, Beavis WD, Belknap JK, Bennett B, Berrettini W, Bleich A, Bogue M, Broman KW, Buck KJ, Buckler E, Burmeister M, Chesler EJ, Cheverud JM, Clapcote S, Cook MN, Cox RD, Crabbe JC, Crusio WE, Darvasi A, Deschepper CF, Doerge RW, Farber CR, Forejt J, Gaile D, Garlow SJ, Geiger H, Gershenfeld H, Gordon T, Gu J, Gu W, de Haan G, Hayes NL, Heller C, Himmelbauer H, Hitzemann R, Hunter K, Hsu HC, Iraqi FA, Ivandic B, Jacob HJ, Jansen RC, Jepsen KJ, Johnson DK, Johnson TE, Kempermann G, Kendziorski C, Kotb M, Kooy RF, Llamas B, Lammert F, Lassalle JM, Lowenstein PR, Lu L, Lusis A, Manly KF, Marcucio R, Matthews D, Medrano JF, Miller DR, Mittleman G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Morris DG, Mott R, Nadeau JH, Nagase H, Nowakowski RS, O’Hara BF, Osadchuk AV, Page GP, Paigen B, Paigen K, Palmer AA, Pan HJ, Peltonen-Palotie L, Peirce J, Pomp D, Pravenec M, Prows DR, Qi Z, Reeves RH, Roder J, Rosen GD, Schadt EE, Schalkwyk LC, Seltzer Z, Shimomura K, Shou S, Sillanpaa MJ, Siracusa LD, Snoeck HW, Spearow JL, Svenson K, Tarantino LM, Threadgill D, Toth LA, Valdar W, de Villena FP, Warden C, Whatley S, Williams RW, Wiltshire T, Yi N, Zhang D, Zhang M, Zou F, Complex Trait C. The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat Genet. 2004; 36:1133-7. [PMID: 15514660]
  22. Shorter JR, Najarian ML, Bell TA, Blanchard M, Ferris MT, Hock P, Kashfeen A, Kirchoff KE, Linnertz CL, Sigmon JS, Miller DR, McMillan L, Pardo-Manuel de Villena F. Whole Genome Sequencing and Progress Toward Full Inbreeding of the Mouse Collaborative Cross Population. G3 (Bethesda). 2019; 9:1303-11. [PMID: 30858237]
  23. Rau CD, Civelek M, Pan C, Lusis AJ. A Suite of Tools for Biologists That Improve Accessibility and Visualization of Large Systems Genetics Datasets: Applications to the Hybrid Mouse Diversity Panel. Methods Mol Biol. 2017; 1488:153-88. [PMID: 27933524]
  24. Ferland RJ, Smith J, Papandrea D, Gracias J, Hains L, Kadiyala SB, O’Brien B, Kang EY, Beyer BS, Herron BJ. Multidimensional Genetic Analysis of Repeated Seizures in the Hybrid Mouse Diversity Panel Reveals a Novel Epileptogenesis Susceptibility Locus. G3 (Bethesda). 2017; 7:2545-58. [PMID: 28620084]
  25. Chintalapudi SR, Maria D, Di Wang X, Bailey JNC. consortium N, International Glaucoma Genetics c, Hysi PG, Wiggs JL, Williams RW, Jablonski MM. Systems genetics identifies a role for Cacna2d1 regulation in elevated intraocular pressure and glaucoma susceptibility. Nat Commun. 2017; 8:1755 [PMID: 29176626]
  26. Kautzman AG, Keeley PW, Ackley CR, Leong S, Whitney IE, Reese BE. Xkr8 Modulates Bipolar Cell Number in the Mouse Retina. Front Neurosci. 2018; 12:876 [PMID: 30559640]
  27. Freeman NE, Templeton JP, Orr WE, Lu L, Williams RW, Geisert EE. Genetic networks in the mouse retina: growth associated protein 43 and phosphatase tensin homolog network. Mol Vis. 2011; 17:1355-72. [PMID: 21655357]
  28. Lu H, Lu L, Williams RW, Jablonski MM. Iris transillumination defect and its gene modulators do not correlate with intraocular pressure in the BXD family of mice. Mol Vis. 2016; 22:224-33. [PMID: 27011731]
  29. Reese BE, Keeley PW. Genomic control of neuronal demographics in the retina. Prog Retin Eye Res. 2016; 55:246-59. [PMID: 27492954]
  30. Whitney IE, Raven MA, Ciobanu DC, Poche RA, Ding Q, Elshatory Y, Gan L, Williams RW, Reese BE. Genetic modulation of horizontal cell number in the mouse retina. Proc Natl Acad Sci USA. 2011; 108:9697-702. [PMID: 21576457]
  31. McDaid AF, Joshi PK, Porcu E, Komljenovic A, Li H, Sorrentino V, Litovchenko M, Bevers RPJ, Rueger S, Reymond A, Bochud M, Deplancke B, Williams RW, Robinson-Rechavi M, Paccaud F, Rousson V, Auwerx J, Wilson JF, Kutalik Z. Bayesian association scan reveals loci associated with human lifespan and linked biomarkers. Nat Commun. 2017; 8:15842 [PMID: 28748955]
  32. Farris SP, Riley BP, Williams RW, Mulligan MK, Miles MF, Lopez MF, Hitzemann R, Iancu OD, Colville A, Walter NAR, Darakjian P, Oberbeck DL, Daunais JB, Zheng CL, Searles RP, McWeeney SK, Grant KA, Mayfield RD. Cross-species molecular dissection across alcohol behavioral domains. Alcohol. 2018; 72:19-31. [PMID: 30213503]
  33. Ashbrook DG, Williams RW, Lu L, Hager R. A cross-species genetic analysis identifies candidate genes for mouse anxiety and human bipolar disorder. Front Behav Neurosci. 2015; 9:171 [PMID: 26190982]
  34. King R, Li Y, Wang J, Struebing FL, Geisert EE. Genomic Locus Modulating IOP in the BXD RI Mouse Strains. G3 (Bethesda). 2018; 8:1571-8. [PMID: 29496776]
  35. Williams RW, Strom RC, Rice DS, Goldowitz D. Genetic and environmental control of variation in retinal ganglion cell number in mice. J Neurosci. 1996; 16:7193-205. [PMID: 8929428]
  36. Heimel JA, Hermans JM, Sommeijer JP. Neuro-Bsik Mouse Phenomics c, Levelt CN. Genetic control of experience-dependent plasticity in the visual cortex. Genes Brain Behav. 2008; 7:915-23. [PMID: 18700840]
  37. King R, Lu L, Williams RW, Geisert EE. Transcriptome networks in the mouse retina: An exon level BXD RI database. Mol Vis. 2015; 21:1235-51. [PMID: 26604663]
  38. Morse HC, , 3rd Chused TM, Hartley JW, Mathieson BJ, Sharrow SO, Taylor BA. Expression of xenotropic murine leukemia viruses as cell-surface gp70 in genetic crosses between strains DBA/2 and C57BL/6. J Exp Med. 1979; 149:1183-96. [PMID: 221612]
  39. Peirce JL, Lu L, Gu J, Silver LM, Williams RW. A new set of BXD recombinant inbred lines from advanced intercross populations in mice. BMC Genet. 2004; 5:7 [PMID: 15117419]
  40. Williams RW, Strom RC, Goldowitz D. Natural variation in neuron number in mice is linked to a major quantitative trait locus on Chr 11. J Neurosci. 1998; 18:138-46. [PMID: 9412494]
  41. Struebing FL, King R, Li Y, Chrenek MA, Lyuboslavsky PN, Sidhu CS, Iuvone PM, Geisert EE. Transcriptional Changes in the Mouse Retina after Ocular Blast Injury: A Role for the Immune System. J Neurotrauma. 2018; 35:118-29. [PMID: 28599600]
  42. Anderson MG, Smith RS, Hawes NL, Zabaleta A, Chang B, Wiggs JL, John SW. Mutations in genes encoding melanosomal proteins cause pigmentary glaucoma in DBA/2J mice. Nat Genet. 2002; 30:81-5. [PMID: 11743578]
  43. Anderson MG, Libby RT, Mao M, Cosma IM, Wilson LA, Smith RS, John SW. Genetic context determines susceptibility to intraocular pressure elevation in a mouse pigmentary glaucoma. BMC Biol. 2006; 4:20 [PMID: 16827931]
  44. Howell GR, Libby RT, Marchant JK, Wilson LA, Cosma IM, Smith RS, Anderson MG, John SW. Absence of glaucoma in DBA/2J mice homozygous for wild-type versions of Gpnmb and Tyrp1. BMC Genet. 2007; 8:45 [PMID: 17608931]
  45. Vázquez-Chona FR, Lu L, Williams RW, Geisert EE. Genomic loci modulating the retinal transcriptome in wound healing. Gene Regul Syst Bio. 2008; 1:327-48. [PMID: 19936100]
  46. Vázquez-Chona FR, Geisert EE. Networks modulating the retinal response to injury: insights from microarrays, expression genetics, and bioinformatics. Adv Exp Med Biol. 2012; 723:649-56. [PMID: 22183389]
  47. Wang J, Geisert EE, Struebing FL. RNA sequencing profiling of the retina in C57BL/6J and DBA/2J mice: Enhancing the retinal microarray data sets from GeneNetwork. Mol Vis. 2019; 25:345-58. [PMID: 31354228]
  48. Struebing FL, Lee RK, Williams RW, Geisert EE. Genetic Networks in Mouse Retinal Ganglion Cells. Front Genet. 2016; 7:169 [PMID: 27733864]
  49. Struebing FL, King R, Li Y, Cooke Bailey JN. consortium N, Wiggs JL, Geisert EE. Genomic loci modulating retinal ganglion cell death following elevated IOP in the mouse. Exp Eye Res. 2018; 169:61-7. [PMID: 29421330]
  50. Ahmed F, Brown KM, Stephan DA, Morrison JC, Johnson EC, Tomarev SI. Microarray analysis of changes in mRNA levels in the rat retina after experimental elevation of intraocular pressure. Invest Ophthalmol Vis Sci. 2004; 45:1247-58. [PMID: 15037594]
  51. Steele MR, Inman DM, Calkins DJ, Horner PJ, Vetter ML. Microarray analysis of retinal gene expression in the DBA/2J model of glaucoma. Invest Ophthalmol Vis Sci. 2006; 47:977-85. [PMID: 16505032]
  52. Fan W, Li X, Wang W, Mo JS, Kaplan H, Cooper NG. Early Involvement of Immune/Inflammatory Response Genes in Retinal Degeneration in DBA/2J Mice. Ophthalmol Eye Dis. 2010; 1:23-41. [PMID: 20352036]
  53. Howell GR, Macalinao DG, Sousa GL, Walden M, Soto I, Kneeland SC, Barbay JM, King BL, Marchant JK, Hibbs M, Stevens B, Barres BA, Clark AF, Libby RT, John SW. Molecular clustering identifies complement and endothelin induction as early events in a mouse model of glaucoma. J Clin Invest. 2011; 121:1429-44. [PMID: 21383504]
  54. Zhou G, Williams RW. Eye1 and Eye2: gene loci that modulate eye size, lens weight, and retinal area in the mouse. Invest Ophthalmol Vis Sci. 1999; 40:817-25. [PMID: 10102277]
  55. Seecharan DJ, Kulkarni AL, Lu L, Rosen GD, Williams RW. Genetic control of interconnected neuronal populations in the mouse primary visual system. J Neurosci. 2003; 23:11178-88. [PMID: 14657177]
  56. Whitney IE, Raven MA, Ciobanu DC, Williams RW, Reese BE. Multiple genes on chromosome 7 regulate dopaminergic amacrine cell number in the mouse retina. Invest Ophthalmol Vis Sci. 2009; 50:1996-2003. [PMID: 19168892]
  57. Whitney IE, Raven MA, Lu L, Williams RW, Reese BE. A QTL on chromosome 10 modulates cone photoreceptor number in the mouse retina. Invest Ophthalmol Vis Sci. 2011; 52:3228-36. [PMID: 21330668]
  58. Munguba GC, Geisert EE, Williams RW, Tapia ML, Lam DK, Bhattacharya SK, Lee RK. Effects of glaucoma on Chrna6 expression in the retina. Curr Eye Res. 2013; 38:150-7. [PMID: 23002780]
  59. Engelbrecht B. The mystery of suffering. Med Law. 1989; 7:635-6. [PMID: 2495402]
  60. Uraoka Z, Akano N, Maki J. Infantile hereditary nephropathies. Nippon Jinzo Gakkai Shi. 1983; 25:888-9. [PMID: 6663874]
  61. Kautzman AG, Keeley PW, Borhanian S, Ackley CR, Reese BE. Genetic Control of Rod Bipolar Cell Number in the Mouse Retina. Front Neurosci. 2018; 12:285 [PMID: 29867309]
  62. Wang J, Li Y, King R, Struebing FL, Geisert EE. Optic nerve regeneration in the mouse is a complex trait modulated by genetic background. Mol Vis. 2018; 24:174-86. [PMID: 29463955]
  63. Gordon MO, Beiser JA, Brandt JD, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, Parrish RK, , 2nd Wilson MR, Kass MA. The Ocular Hypertension Treatment Study: baseline factors that predict the onset of primary open-angle glaucoma. Arch Ophthalmol. 2002; 120:714-20.-, discussion 829–30.. [PMID: 12049575]
  64. Quigley HA. Number of people with glaucoma worldwide. Br J Ophthalmol. 1996; 80:389-93. [PMID: 8695555]
  65. Thylefors B, Negrel AD. The global impact of glaucoma. Bull World Health Organ. 1994; 72:323-6. [PMID: 8062393]
  66. Leske MC, Heijl A, Hyman L, Bengtsson B, Dong L, Yang Z, Group E. Predictors of long-term progression in the early manifest glaucoma trial. Ophthalmology. 2007; 114:1965-72. [PMID: 17628686]
  67. Liu Y, Allingham RR. Molecular genetics in glaucoma. Exp Eye Res. 2011; 93:331-9. [PMID: 21871452]
  68. Springelkamp H, Iglesias AI, Mishra A, Hohn R, Wojciechowski R, Khawaja AP, Nag A, Wang YX, Wang JJ, Cuellar-Partida G, Gibson J, Cooke Bailey JN, Vithana EN, Gharahkhani P, Boutin T, Ramdas WD, Zeller T, Luben RN, Yonova-Doing E, Viswanathan AC, Yazar S, Cree AJ, Haines JL, Koh JY, Souzeau E, Wilson JF, Amin N, Muller C, Venturini C, Kearns LS, Hee Kang J, Consortium N, Tham YC, Zhou T, van Leeuwen EM, Nickels S, Sanfilippo P, Liao J, Linde HV, Zhao W, van Koolwijk LM, Zheng L, Rivadeneira F, Baskaran M, van der Lee SJ, Perera S, de Jong PT, Oostra BA, Uitterlinden AG, Fan Q, Hofman A, Shyong Tai E, Vingerling JR, Sim X, Wolfs RC, Teo YY, Lemij HG, Khor CC, Willemsen R, Lackner KJ, Aung T, Jansonius NM, Montgomery G, Wild PS, Young TL, Burdon KP, Hysi PG, Pasquale LR, Wong TY, Klaver CC, Hewitt AW, Jonas JB, Mitchell P, Lotery AJ, Foster PJ, Vitart V, Pfeiffer N, Craig JE, Mackey DA, Hammond CJ, Wiggs JL, Cheng CY, van Duijn CM, MacGregor S. New insights into the genetics of primary open-angle glaucoma based on meta-analyses of intraocular pressure and optic disc characteristics. Hum Mol Genet. 2017; 26:438-53 [PMID: 28073927]
  69. Nickells RW. The cell and molecular biology of glaucoma: mechanisms of retinal ganglion cell death. Invest Ophthalmol Vis Sci. 2012; 53:2476-81. [PMID: 22562845]
  70. Herndon LW, Weizer JS, Stinnett SS. Central corneal thickness as a risk factor for advanced glaucoma damage. Arch Ophthalmol. 2004; 122:17-21. [PMID: 14718289]
  71. Klein BE, Klein R, Lee KE. Heritability of risk factors for primary open-angle glaucoma: the Beaver Dam Eye Study. Invest Ophthalmol Vis Sci. 2004; 45:59-62. [PMID: 14691154]
  72. Stone EM, Fingert JH, Alward WL, Nguyen TD, Polansky JR, Sunden SL, Nishimura D, Clark AF, Nystuen A, Nichols BE, Mackey DA, Ritch R, Kalenak JW, Craven ER, Sheffield VC. Identification of a gene that causes primary open angle glaucoma. Science. 1997; 275:668-70. [PMID: 9005853]
  73. Wiggs JL. Genetic etiologies of glaucoma. Arch Ophthalmol. 2007; 125:30-7. [PMID: 17210849]
  74. Medeiros FA, Sample PA, Weinreb RN. Corneal thickness measurements and visual function abnormalities in ocular hypertensive patients. Am J Ophthalmol. 2003; 135:131-7. [PMID: 12566014]
  75. European Glaucoma Prevention Study G. Miglior S, Pfeiffer N, Torri V, Zeyen T, Cunha-Vaz J, Adamsons I. Predictive factors for open-angle glaucoma among patients with ocular hypertension in the European Glaucoma Prevention Study. Ophthalmology. 2007; 114:3-9. [PMID: 17070596]
  76. Struebing FL, Geisert EE. What Animal Models Can Tell Us About Glaucoma. Prog Mol Biol Transl Sci. 2015; 134:365-80. [PMID: 26310165]
  77. John SW, Smith RS, Savinova OV, Hawes NL, Chang B, Turnbull D, Davisson M, Roderick TH, Heckenlively JR. Essential iris atrophy, pigment dispersion, and glaucoma in DBA/2J mice. Invest Ophthalmol Vis Sci. 1998; 39:951-62. [PMID: 9579474]
  78. Williams PA, Harder JM, Foxworth NE, Cardozo BH, Cochran KE, John SWM. Nicotinamide and WLD(S) Act Together to Prevent Neurodegeneration in Glaucoma. Front Neurosci. 2017; 11:232 [PMID: 28487632]
  79. Boussommier-Calleja A, Li G, Wilson A, Ziskind T, Scinteie OE, Ashpole NE, Sherwood JM, Farsiu S, Challa P, Gonzalez P, Downs JC, Ethier CR, Stamer WD, Overby DR. Physical Factors Affecting Outflow Facility Measurements in Mice. Invest Ophthalmol Vis Sci. 2015; 56:8331-9. [PMID: 26720486]
  80. Stamer WD, Braakman ST, Zhou EH, Ethier CR, Fredberg JJ, Overby DR, Johnson M. Biomechanics of Schlemm’s canal endothelium and intraocular pressure reduction. Prog Retin Eye Res. 2015; 44:86-98. [PMID: 25223880]
  81. Springelkamp H, Hohn R, Mishra A, Hysi PG, Khor CC, Loomis SJ, Bailey JN, Gibson J, Thorleifsson G, Janssen SF, Luo X, Ramdas WD, Vithana E, Nongpiur ME, Montgomery GW, Xu L, Mountain JE, Gharahkhani P, Lu Y, Amin N, Karssen LC, Sim KS, van Leeuwen EM, Iglesias AI, Verhoeven VJ, Hauser MA, Loon SC, Despriet DD, Nag A, Venturini C, Sanfilippo PG, Schillert A, Kang JH, Landers J, Jonasson F, Cree AJ, van Koolwijk LM, Rivadeneira F, Souzeau E, Jonsson V, Menon G. Blue Mountains Eye Study Gg, Weinreb RN, de Jong PT, Oostra BA, Uitterlinden AG, Hofman A, Ennis S, Thorsteinsdottir U, Burdon KP, Consortium N, Wellcome Trust Case Control C, Spector TD, Mirshahi A, Saw SM, Vingerling JR, Teo YY, Haines JL, Wolfs RC, Lemij HG, Tai ES, Jansonius NM, Jonas JB, Cheng CY, Aung T, Viswanathan AC, Klaver CC, Craig JE, Macgregor S, Mackey DA, Lotery AJ, Stefansson K, Bergen AA, Young TL, Wiggs JL, Pfeiffer N, Wong TY, Pasquale LR, Hewitt AW, van Duijn CM, Hammond CJ. Meta-analysis of genome-wide association studies identifies novel loci that influence cupping and the glaucomatous process. Nat Commun. 2014; 5:4883 [PMID: 25241763]
  82. Nag A, Venturini C, Small KS. International Glaucoma Genetics C, Young TL, Viswanathan AC, Mackey DA, Hysi PG, Hammond C. A genome-wide association study of intra-ocular pressure suggests a novel association in the gene FAM125B in the TwinsUK cohort. Hum Mol Genet. 2014; 23:3343-8. [PMID: 24518671]
  83. Chen F, Klein AP, Klein BE, Lee KE, Truitt B, Klein R, Iyengar SK, Duggal P. Exome array analysis identifies CAV1/CAV2 as a susceptibility locus for intraocular pressure. Invest Ophthalmol Vis Sci. 2014; 56:544-51. [PMID: 25525164]
  84. Choquet H, Thai KK, Yin J, Hoffmann TJ, Kvale MN, Banda Y, Schaefer C, Risch N, Nair KS, Melles R, Jorgenson E. A large multi-ethnic genome-wide association study identifies novel genetic loci for intraocular pressure. Nat Commun. 2017; 8:2108 [PMID: 29235454]
  85. Ozel AB, Moroi SE, Reed DM, Nika M, Schmidt CM, Akbari S, Scott K, Rozsa F, Pawar H, Musch DC, Lichter PR, Gaasterland D, Branham K, Gilbert J, Garnai SJ, Chen W, Othman M, Heckenlively J, Swaroop A, Abecasis G, Friedman DS, Zack D, Ashley-Koch A, Ulmer M, Kang JH, Consortium N, Liu Y, Yaspan BL, Haines J, Allingham RR, Hauser MA, Pasquale L, Wiggs J, Richards JE, Li JZ. Genome-wide association study and meta-analysis of intraocular pressure. Hum Genet. 2014; 133:41-57. [PMID: 24002674]
  86. Springelkamp H, Iglesias AI, Mishra A, Hohn R, Wojciechowski R, Khawaja AP, Nag A, Wang YX, Wang JJ, Cuellar-Partida G, Gibson J, Bailey JN, Vithana EN, Gharahkhani P, Boutin T, Ramdas WD, Zeller T, Luben RN, Yonova-Doing E, Viswanathan AC, Yazar S, Cree AJ, Haines JL, Koh JY, Souzeau E, Wilson JF, Amin N, Muller C, Venturini C, Kearns LS, Kang JH, Consortium N, Tham YC, Zhou T, van Leeuwen EM, Nickels S, Sanfilippo P, Liao J, van der Linde H, Zhao W, van Koolwijk LM, Zheng L, Rivadeneira F, Baskaran M, van der Lee SJ, Perera S, de Jong PT, Oostra BA, Uitterlinden AG, Fan Q, Hofman A, Tai ES, Vingerling JR, Sim X, Wolfs RC, Teo YY, Lemij HG, Khor CC, Willemsen R, Lackner KJ, Aung T, Jansonius NM, Montgomery G, Wild PS, Young TL, Burdon KP, Hysi PG, Pasquale LR, Wong TY, Klaver CC, Hewitt AW, Jonas JB, Mitchell P, Lotery AJ, Foster PJ, Vitart V, Pfeiffer N, Craig JE, Mackey DA, Hammond CJ, Wiggs JL, Cheng CY, van Duijn CM, MacGregor S. New insights into the genetics of primary open-angle glaucoma based on meta-analyses of intraocular pressure and optic disc characteristics. Hum Mol Genet. 2017; 26:438-53. [PMID: 28073927]
  87. MacGregor S, Ong JS, An J, Han X, Zhou T, Siggs OM, Law MH, Souzeau E, Sharma S, Lynn DJ, Beesley J, Sheldrick B, Mills RA, Landers J, Ruddle JB, Graham SL, Healey PR, White AJR, Casson RJ, Best S, Grigg JR, Goldberg I, Powell JE, Whiteman DC, Radford-Smith GL, Martin NG, Montgomery GW, Burdon KP, Mackey DA, Gharahkhani P, Craig JE, Hewitt AW. Genome-wide association study of intraocular pressure uncovers new pathways to glaucoma. Nat Genet. 2018; 50:1067-71. [PMID: 30054594]
  88. Khawaja AP, Cooke Bailey JN, Wareham NJ, Scott RA, Simcoe M, Igo RP, , Jr Song YE, Wojciechowski R, Cheng CY, Khaw PT, Pasquale LR, Haines JL, Foster PJ, Wiggs JL, Hammond CJ, Hysi PG, Eye UKB, Vision C, Consortium N. Genome-wide analyses identify 68 new loci associated with intraocular pressure and improve risk prediction for primary open-angle glaucoma. Nat Genet. 2018; 50:778-82. [PMID: 29785010]
  89. Choquet H, Paylakhi S, Kneeland SC, Thai KK, Hoffmann TJ, Yin J, Kvale MN, Banda Y, Tolman NG, Williams PA, Schaefer C, Melles RB, Risch N, John SWM, Nair KS, Jorgenson E. A multiethnic genome-wide association study of primary open-angle glaucoma identifies novel risk loci. Nat Commun. 2018; 9:2278 [PMID: 29891935]
  90. Carnes MU, Allingham RR, Ashley-Koch A, Hauser MA. Transcriptome analysis of adult and fetal trabecular meshwork, cornea, and ciliary body tissues by RNA sequencing. Exp Eye Res. 2018; 167:91-9. [PMID: 27914989]
  91. Craig JE, Han X, Qassim A, Hassall M, Cooke Bailey JN, Kinzy TG, Khawaja AP, An J, Marshall H, Gharahkhani P, Igo RP, , Jr Graham SL, Healey PR, Ong JS, Zhou T, Siggs O, Law MH, Souzeau E, Ridge B, Hysi PG, Burdon KP, Mills RA, Landers J, Ruddle JB, Agar A, Galanopoulos A, White AJR, Willoughby CE, Andrew NH, Best S, Vincent AL, Goldberg I, Radford-Smith G, Martin NG, Montgomery GW, Vitart V, Hoehn R, Wojciechowski R, Jonas JB, Aung T, Pasquale LR, Cree AJ, Sivaprasad S, Vallabh NA. consortium N, Eye UKB, Vision C, Viswanathan AC, Pasutto F, Haines JL, Klaver CCW, van Duijn CM, Casson RJ, Foster PJ, Khaw PT, Hammond CJ, Mackey DA, Mitchell P, Lotery AJ, Wiggs JL, Hewitt AW, MacGregor S. Multitrait analysis of glaucoma identifies new risk loci and enables polygenic prediction of disease susceptibility and progression. Nat Genet. 2020; 52:160-6. [PMID: 31959993]
  92. Oda H, Takeichi M. Evolution: structural and functional diversity of cadherin at the adherens junction. J Cell Biol. 2011; 193:1137-46. [PMID: 21708975]
  93. Jontes JD. The Cadherin Superfamily in Neural Circuit Assembly. Cold Spring Harb Perspect Biol. 2018; 10 [PMID: 28778868]
  94. Gul IS, Hulpiau P, Saeys Y, van Roy F. Evolution and diversity of cadherins and catenins. Exp Cell Res. 2017; 358:3-9. [PMID: 28268172]
  95. O’Callaghan J, Crosbie DE, Cassidy PS, Sherwood JM, Flugel-Koch C, Lutjen-Drecoll E, Humphries MM, Reina-Torres E, Wallace D, Kiang AS, Campbell M, Stamer WD, Overby DR, O’Brien C, Tam LCS, Humphries P. Therapeutic potential of AAV-mediated MMP-3 secretion from corneal endothelium in treating glaucoma. Hum Mol Genet. 2017; 26:1230-46. [PMID: 28158775]
  96. Tam LC, Reina-Torres E, Sherwood JM, Cassidy PS, Crosbie DE, Lutjen-Drecoll E, Flugel-Koch C, Perkumas K, Humphries MM, Kiang AS, O’Callaghan J, Callanan JJ, Read AT, Ethier CR, O’Brien C, Lawrence M, Campbell M, Stamer WD, Overby DR, Humphries P. Enhancement of Outflow Facility in the Murine Eye by Targeting Selected Tight-Junctions of Schlemm’s Canal Endothelia. Sci Rep. 2017; 7:40717 [PMID: 28091584]
  97. Wagner AH, Anand VN, Wang WH, Chatterton JE, Sun D, Shepard AR, Jacobson N, Pang IH, Deluca AP, Casavant TL, Scheetz TE, Mullins RF, Braun TA, Clark AF. Exon-level expression profiling of ocular tissues. Exp Eye Res. 2013; 111:105-11. [PMID: 23500522]
  98. Wecker T, Han H, Borner J, Grehn F, Schlunck G. Effects of TGF-beta2 on cadherins and beta-catenin in human trabecular meshwork cells. Invest Ophthalmol Vis Sci. 2013; 54:6456-62. [PMID: 24003087]
  99. Webber HC, Bermudez JY, Millar JC, Mao W, Clark AF. The Role of Wnt/beta-Catenin Signaling and K-Cadherin in the Regulation of Intraocular Pressure. Invest Ophthalmol Vis Sci. 2018; 59:1454-66. [PMID: 29625468]
  100. Tipold A, Zurbriggen A, Moore P, Schijns V, Jungi TW. Generation and functional characterisation of canine bone marrow-derived macrophages. Res Vet Sci. 1998; 64:125-32. [PMID: 9625468]
  101. McDowell CM, Tebow HE, Wordinger RJ, Clark AF. Smad3 is necessary for transforming growth factor-beta2 induced ocular hypertension in mice. Exp Eye Res. 2013; 116:419-23. [PMID: 24184030]
  102. Rodriguez AR, de Sevilla Muller LP, Brecha NC. The RNA binding protein RBPMS is a selective marker of ganglion cells in the mammalian retina. J Comp Neurol. 2014; 522:1411-43. [PMID: 24318667]
  103. Brocker C, Lassen N, Estey T, Pappa A, Cantore M, Orlova VV, Chavakis T, Kavanagh KL, Oppermann U, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem. 2010; 285:18452-63. [PMID: 20207735]
  104. Brocker C, Cantore M, Failli P, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) attenuates reactive aldehyde and oxidative stress induced cytotoxicity. Chem Biol Interact. 2011; 191:269-77. [PMID: 21338592]
  105. Wong JW, Chan CL, Tang WK, Cheng CH, Fong WP. Is antiquitin a mitochondrial Enzyme? J Cell Biochem. 2010; 109:74-81. [PMID: 19885858]
  106. Lunn ER, Perry VH, Brown MC, Rosen H, Gordon S. Absence of Wallerian Degeneration does not Hinder Regeneration in Peripheral Nerve. Eur J Neurosci. 1989; 1:27-33. [PMID: 12106171]
  107. Perry VH, Brown MC, Lunn ER. Very Slow Retrograde and Wallerian Degeneration in the CNS of C57BL/Ola Mice. Eur J Neurosci. 1991; 3:102-5. [PMID: 12106273]
  108. Wang JT, Medress ZA, Vargas ME, Barres BA. Local axonal protection by WldS as revealed by conditional regulation of protein stability. Proc Natl Acad Sci USA. 2015; 112:10093-100. [PMID: 26209654]
  109. Zamorano B. Subcellular localization of prostaglandin-E2 in rat heart tissue. Cardiovasc Drugs Ther. 1991; 5:655-7. [PMID: 1878336]
  110. Williams PA, Harder JM, John SWM. Glaucoma as a Metabolic Optic Neuropathy: Making the Case for Nicotinamide Treatment in Glaucoma. J Glaucoma. 2017; 26:1161-8. [PMID: 28858158]
  111. Toh T, Liew SH, MacKinnon JR, Hewitt AW, Poulsen JL, Spector TD, Gilbert CE, Craig JE, Hammond CJ, Mackey DA. Central corneal thickness is highly heritable: the twin eye studies. Invest Ophthalmol Vis Sci. 2005; 46:3718-22. [PMID: 16186354]
  112. Zheng Y, Ge J, Huang G, Zhang J, Liu B, Hur YM, He M. Heritability of central corneal thickness in Chinese: the Guangzhou Twin Eye Study. Invest Ophthalmol Vis Sci. 2008; 49:4303-7. [PMID: 18502994]
  113. Dimasi DP, Burdon KP, Craig JE. The genetics of central corneal thickness. Br J Ophthalmol. 2010; 94:971-6. [PMID: 19556215]
  114. Medeiros FA, Sample PA, Zangwill LM, Bowd C, Aihara M, Weinreb RN. Corneal thickness as a risk factor for visual field loss in patients with preperimetric glaucomatous optic neuropathy. Am J Ophthalmol. 2003; 136:805-13. [PMID: 14597030]
  115. Wiggs JL, Hauser MA, Abdrabou W, Allingham RR, Budenz DL, Delbono E, Friedman DS, Kang JH, Gaasterland D, Gaasterland T, Lee RK, Lichter PR, Loomis S, Liu Y, McCarty C, Medeiros FA, Moroi SE, Olson LM, Realini A, Richards JE, Rozsa FW, Schuman JS, Singh K, Stein JD, Vollrath D, Weinreb RN, Wollstein G, Yaspan BL, Yoneyama S, Zack D, Zhang K, Pericak-Vance M, Pasquale LR, Haines JL. The NEIGHBOR consortium primary open-angle glaucoma genome-wide association study: rationale, study design, and clinical variables. J Glaucoma. 2013; 22:517-25. [PMID: 22828004]
  116. Springelkamp H, Mishra A, Hysi PG, Gharahkhani P, Hohn R, Khor CC, Cooke Bailey JN, Luo X, Ramdas WD, Vithana E, Koh V, Yazar S, Xu L, Forward H, Kearns LS, Amin N, Iglesias AI, Sim KS, van Leeuwen EM, Demirkan A, van der Lee S, Loon SC, Rivadeneira F, Nag A, Sanfilippo PG, Schillert A, de Jong PT, Oostra BA, Uitterlinden AG, Hofman A, Consortium N, Zhou T, Burdon KP, Spector TD, Lackner KJ, Saw SM, Vingerling JR, Teo YY, Pasquale LR, Wolfs RC, Lemij HG, Tai ES, Jonas JB, Cheng CY, Aung T, Jansonius NM, Klaver CC, Craig JE, Young TL, Haines JL, MacGregor S, Mackey DA, Pfeiffer N, Wong TY, Wiggs JL, Hewitt AW, van Duijn CM, Hammond CJ. Meta-analysis of Genome-Wide Association Studies Identifies Novel Loci Associated With Optic Disc Morphology. Genet Epidemiol. 2015; 39:207-16. [PMID: 25631615]
  117. Bailey JN, Loomis SJ, Kang JH, Allingham RR, Gharahkhani P, Khor CC, Burdon KP, Aschard H, Chasman DI, Igo RP, , Jr Hysi PG, Glastonbury CA, Ashley-Koch A, Brilliant M, Brown AA, Budenz DL, Buil A, Cheng CY, Choi H, Christen WG, Curhan G, De Vivo I, Fingert JH, Foster PJ, Fuchs C, Gaasterland D, Gaasterland T, Hewitt AW, Hu F, Hunter DJ, Khawaja AP, Lee RK, Li Z, Lichter PR, Mackey DA, McGuffin P, Mitchell P, Moroi SE, Perera SA, Pepper KW, Qi Q, Realini T, Richards JE, Ridker PM, Rimm E, Ritch R, Ritchie M, Schuman JS, Scott WK, Singh K, Sit AJ, Song YE, Tamimi RM, Topouzis F, Viswanathan AC, Verma SS, Vollrath D, Wang JJ, Weisschuh N, Wissinger B, Wollstein G, Wong TY, Yaspan BL, Zack DJ, Zhang K, Study EN, Consortium A, Weinreb RN, Pericak-Vance MA, Small K, Hammond CJ, Aung T, Liu Y, Vithana EN, MacGregor S, Craig JE, Kraft P, Howell G, Hauser MA, Pasquale LR, Haines JL, Wiggs JL. Genome-wide association analysis identifies TXNRD2, ATXN2 and FOXC1 as susceptibility loci for primary open-angle glaucoma. Nat Genet. 2016; 48:189-94. [PMID: 26752265]
  118. Zhou H, Yoshioka T, Nathans J. Retina-derived POU-domain factor-1: a complex POU-domain gene implicated in the development of retinal ganglion and amacrine cells. J Neurosci. 1996; 16:2261-74. [PMID: 8601806]
  119. Keele GR, Crouse WL, Kelada SNP, Valdar W. Determinants of QTL Mapping Power in the Realized Collaborative Cross. G3 (Bethesda). 2019; 9:1707-27. [PMID: 30914424]