Molecular Vision 2010; 16:2007-2015 <http://www.molvis.org/molvis/v16/a216>
Received 13 August 2010 | Accepted 4 October 2010 | Published 8 October 2010

Cat-Map: putting cataract on the map

Alan Shiels,1 Thomas M. Bennett,1 J. Fielding Hejtmancik2

1Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO; 2Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda MD

Correspondence to: Dr. Alan Shiels, Department of Ophthalmology and Visual Sciences, Campus Box 8096, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110; Phone: (314) 362-1637; FAX: (314) 362-1646; email: shiels@vision.wustl.edu

Abstract

Lens opacities, or cataract(s), may be inherited as a classic Mendelian disorder usually with early-onset or, more commonly, acquired with age as a multi-factorial or complex trait. Many genetic forms of cataract have been described in mice and other animal models. Considerable progress has been made in mapping and identifying the genes and mutations responsible for inherited forms of cataract, and genetic determinants of age-related cataract are beginning to be discovered. To provide a convenient and accurate summary of current information focused on the increasing genetic complexity of Mendelian and age-related cataract we have created an online chromosome map and reference database for cataract in humans and mice (Cat-Map).

1. Introduction

Cataract is a light-scattering disorder of the crystalline lens that despite surgical treatment remains an important cause of visual impairment in the United States and worldwide [1,2]. Typically, cataract is acquired with age (>50 years) as a multi-factorial disorder involving complex interactions between genetic and environmental risk factors [3]. In some cases however, cataract may be inherited as a classic Mendelian disorder (~1/10,000 births), either in association with other ocular and/or systemic abnormalities or, as an isolated lens phenotype [4,5]. By contrast with age-related cataract, Mendelian forms of cataract can occur at any age, however, most have an early-onset either presenting at birth (congenital), during infancy, childhood or adolescence. Beyond age-at-onset both inherited and age-related forms of cataract are clinically heterogeneous with respect to location, size, shape, density and even color of opacity within the lens [6-8].

In 1968 an inherited form of isolated cataract, which had previously been linked to the Duffy blood group locus, became the first monogenic disorder in humans to be assigned to an autosome (chromosome 1) [9]. Since then, considerable progress has been made in mapping and identifying genes for inherited cataract mostly by linkage analysis in extended families. All three classical types of Mendelian inheritance have been described for familial cataract; however, the existence of Y-linked cataract remains to be confirmed [10]. Similarly, genetic forms of cataract are also found in laboratory mice, and many spontaneous, chemical or radiation induced, and genetically engineered (transgenic and gene-targeted) mutant strains have been described [11]. Many of the causative genes in mice are orthologs of those identified in humans, and these mutant strains provide valuable model systems to investigate human cataract.

In contrast to Mendelian cataract our understanding of the genetic determinants of complex age-related cataract is less well advanced. Candidate gene association studies are providing increasing evidence that variations in some of the genes linked with inherited forms of early-onset cataract are associated with the much more common forms of age-related cataract. However, the advent of genome-wide association studies predicts that the genetic diversity of age-related cataract will likely extend beyond known genes for inherited cataract.

In an effort to aid access to genetic information focused on cataract we have created an online chromosome map and reference database for inherited and age-related forms of cataract in humans and mice (Cat-Map). Genes and loci linked or associated with familial and acquired forms of primary cataract, respectively, were obtained by keyword searches of PubMed, Online Mendelian Inheritance in Man (OMIM), and other databases accessible through the National Center for Biotechnology Information (NCBI) website [12]. Inheritance pattern, family or population origin, gene mutation or variation, cataract appearance, and any associated ocular or systemic phenotypes were also noted. Gene mutations and variations were numbered according to standard nomenclature recommendations starting with the A of the ATG start-codon [13]. Genes and loci for syndromes with cataract and mouse mutants with cataract were also included. Data for each chromosome were formatted in Microsoft Excel and links provided to the EntrezGene and PubMed databases. Due to the need for regular updating of content we have not included chromosome tables with the present text. The most recent updates can be accessed at the Cat-Map website. Here we provide a brief overview of the genes underlying Mendelian and age-related forms of cataract in humans and mice.

2. Genes for Mendelian cataract

Mendelian forms of cataract are highly clinically heterogeneous and comprise a broad spectrum of genetic conditions. In a clinical setting it is pragmatic to distinguish between “syndromic” and “non-syndromic” forms of cataract based on the presence or absence of associated ocular and/or systemic abnormalities, respectively. However, it should be noted that these are relative, rather than precise, clinical terms essentially describing opposite phenotypic extremes of the cataract spectrum disorders. Some clinical overlap between syndromic and non-syndromic forms of cataract is inevitable largely because different mutations in a single gene can exhibit pleiotropic effects that result in distinct and seemingly unrelated phenotypes with variable onset, severity and course.

2.1 Genes for non-syndromic cataract-- Non-syndromic forms of familial cataract present as an isolated or primary lens disorder in the absence of other clinically prominent phenotypes. Under slit-lamp examination non-syndromic familial cataract exhibits a diverse range of lens opacities including, total, nuclear, lamellar, sutural, and anterior/posterior polar or sub-capsular, however, there is no universally accepted classification system [6,7]. Non-syndromic cataract also exhibits considerable inter- and intra-familial variation affecting age-at-onset, rate of progression, and post-surgical visual acuity. Moreover, relatively subtle ocular signs occasionally accompany primary cataract further complicating clinical classification. These pleiotropic effects may involve the anterior segment (e.g., microcornea), refractive error (e.g., myopia), and eye movement disorders (nystagmus, strabismus, amblyopia); further highlighting the importance of the lens in normal eye development and refractive vision. Non-syndromic forms of cataract may be grouped based on the known underlying genes, which currently include those coding for crystallins, membrane/cytoskeleton proteins and a transcription factor.

2.1.1 Crystallin genes-- Around 60 different mutations segregating in some 98 families have been identified in 10 human crystallin genes, including those coding for both α-crystallins (CRYAA, 21q; CRYAB 11q), two acidic β-crystallins (CRYBA1, 17q; CRYBA4, 22q), three basic β-crystallins (CRYBB1, CRYBB2, and CRYBB3 all on 22q), and three γ-crystallins (CRYGC, 2q; CRYGD, 2q; and CRYGS, 3q). Generally, mutations in crystallin genes tend to cause nuclear or lamellar lens opacities. In addition to primary cataract, mutations in crystallin genes, particularly those in CRYAA, may be associated with microcornea. However, a noteworthy exception is that mutations in CRYAB may underlie cataract and/or myopathy phenotypes [4,5]. Overall, crystallin mutations, mostly missense, account for about 50% of non-syndromic familial cataract reported so far.

Several unusually recurrent mutations in crystallin genes are noteworthy. First, a p.P24T missense substitution in CRYGD has been reported in eight families, and a p.P24S missense substitution has been shown to reflect the ancestral protein sequence found in non-primate mammals [14]. Second, the apparent p.Q155X nonsense mutation in CRYBB2 in fact results from a gene conversion event with the adjacent CRYBB2 pseudogene [15]. Interestingly, a rare case of a severely affected family member homozygous for the p.Q155X conversion mutation developed microphthalmia, microcornea, leukocoria, congenital nystagmus, and a dysplastic lens [16]. Third, a splice (g.IVS+1G>A/C) or a deletion (p.G91del) mutation segregating in each of five families has been detected in CRYBA1 (17q), which encodes two acidic-beta crystallins (CRYBA3 and CRYBA1). Finally, recurrent arginine substitutions are common in CRYAA, CRYGD, and CRYGC [4,5].

In cataract mouse strains, over 30 spontaneous or induced mutations have been reported in ten crystallin genes. Over half of these strains carry mutations in six crystallin genes (Cryaa, Cryba1, Crybb2, Crygc, Crygd, and Crygs) that are syntenic with human counterparts linked with cataract, thereby providing relevant mouse models for human cataract [11]. The remainder of the crystallin mutant strains, however, serves to highlight species differences between humans and mice. Whereas, all seven Cryg genes harbor mutations in mice (Cryga-f, Crygs); mutations in humans are restricted to CRYGC, CRYGD, and CRYGS. Moreover, two γ-crystallin genes in the mouse (Cryge, Crygf) are represented by pseudo-genes in humans (CRYGEP1, CRYGFP). Finally, mice null for Cryaa and Crybb2, but not Cryab, also develop cataract as do transgenic or knock-in mice harboring several humanized mutations in Cryaa (p.R116C, p.R49C) [17,18].

2.1.2 Genes for membrane or cytoskeleton proteins-- Mutations in at least ten other genes that encode membrane proteins or cytoskeletal proteins have been linked with about 35% of non-syndromic cataract, mostly with autosomal dominant inheritance [4,5]. Mutations in many mouse counterparts of these human genes have also been reported [11].

2.1.2.1 Connexin genes-- The genes for gap-junction proteins (or connexins) alpha-3 (GJA3, 13q) and alpha-8 (GJA8, 1q) harbor over 34 different mutations (mostly missense) segregating in about 38 families. Notably, one of the mutations in GJA8 (p.P88S) underlies the historically important “zonular pulverulent” cataract in an English family that was first linked to chromosome 1 in 1968 [19]. Similarly, a mutation in GJA3 (p.L11S) underlies the unusual “ant-egg” cataract described in a Danish family [20]. Mutations in connexin genes are often associated with pulverulent (dust-like) nuclear opacities and, occasionally, GJA8 mutations are associated with microcornea. Combined, GJA3 and GJA8 mutations account for about 20% of non-syndromic familial cataract. In mice, homozygous loss of Gja3 or Gja8 results in cataract, whereas, heterozygous loss does not [21,22].

2.1.2.2 Genes for major intrinsic proteins-- Mutations in the genes for major intrinsic protein (MIP, 12q), a member of the aquaporin family of water channels, and lens intrinsic membrane protein-2 (LIM2, 19q), a member of the PMP22/claudin family of cell-junction proteins, account for about 5% of familial cataract. All seven mutations in MIP are associated with autosomal dominant cataract, whereas, both mutations in LIM2 underlie autosomal recessive cataract. In mice, heterozygous loss of Mip is sufficient to trigger cataract, whereas, homozygous loss of Lim2 is required for cataract development [23,24].

2.1.2.3 Genes for other membrane-associated proteins-- Several unexpected genes involved in membrane-associated signaling or transport processes are causative for about 5% of non-syndromic familial cataract. These include genes encoding: transmembrane protein-114 (TMEM114, 16p) [25], a sequence homolog of LIM2 with unknown function; chromatin modifying protein-4B or charged multi-vesicular protein-4B (CHMP4B, 20q) [26], a core component of the endosome sorting complex required for transport-III (ESCRT-III); and EPH receptor A2 (EPHA2, 1p) [27-30], a key component of the Eph-ephrin bidirectional signaling pathway. In mice, no mutations in Tmem114 have been reported. Mice null for Epha2 develop cataract, whereas, Chmp4b null mice are embryonic lethal [29,31].

2.1.2.4 Genes for cytoskeleton proteins-- Mutations in the genes for beaded-filament structural protein-1 (BFSP1, 20p) and protein-2 (BFSP2, 3q), which encode intermediate filament-like cytoskeletal proteins, constitute about 4% of familial cataract. A recurrent in-frame deletion mutation has been found in BFSP2 (p.E233del). Recently, missense mutations in the vimentin gene (VIM, 10p) have been associated with cataract in humans and mice [32,33]. Surprisingly, homozygous loss of Vim, Bfsp1, or Bfsp2 is not sufficient to trigger overt cataract in mice, and several commonly used wild-type strains (e.g., 129, FVB) have been found to carry a naturally occurring deletion mutation in Bfsp2 that is associated with a subtle, progressive loss of optical quality [34-38].

2.1.3 Transcription factor genes-- Mutations in the gene for heat-shock transcription factor-4 (HSF4, 16q) underlie about 6% of non-syndromic familial cataract. A missense mutation in HSF4 (p.R119C) segregates in the historically important Marner cataract family [39]. Mice null or mutant for Hsf4 also develop cataract [11]. Whereas, mutations in HSF4 appear to be restricted to an autosomal dominant or recessive cataract phenotype, mutations in several other transcription factors are usually associated with cataract plus other significant ocular defects (see below).

2.2 Genes for syndromic cataract-- Syndromic forms of cataract present as a secondary or variably associated symptom of a genetic syndrome or metabolic disorder that features other defining ocular and/or systemic abnormalities. Under slit-lamp examination syndromic cataract shows variable phenotypes similar to those of non-syndromic opacities (e.g., sutural, posterior sub-capsular) that in some disorders constitute part of the differential diagnosis (e.g., Werner syndrome on 8q, Nance-Horan syndrome on Xp, and Lowe syndrome on Xq). The underlying genetic mechanisms are diverse and include: chromosome abnormalities (e.g., Down syndrome on 21), triplet repeat-disorders (e.g., myotonic dystrophy on 19q), loss-of-heterozygosity (e.g., neurofibromatosis type-2 on 22q), mitochondrial disorders (e.g., myopathy, encephalopathy, lactic acidosis and stroke-like, or MELAS, syndrome), and genetically complex disorders such as diabetes mellitus [40].

Other ocular defects associated with syndromic cataract vary widely affecting the optic nerve, retina, vitreous body, and anterior segment, including; Norrie disease (Xp), gyrate atrophy (10q), optic atrophy-3 (19q), aniridia/Peters anomaly (11p), and Stickler syndrome type-1 (12q). In particular, mutations in several genes for transcription factors including the homeobox genes, PAX6 (11q), FOXE3 (1q), PITX3 (10q) and VSX2 (14q), and the bZIP transcription factor V-MAF avian musculo-aponeurotic fibrosarcoma oncogene homolog (MAF, 16q) underlie cataract plus anterior segment developmental disorders and microphthalmia sometimes associated with secondary glaucoma [41]. In one family with autosomal dominant posterior polar cataract, two siblings homozygous for a deletion mutation in PITX3 exhibited severe microphthalmia and neuro-developmental abnormalities [42]. Mice with mutations in these transcription factor genes also inherit significant ocular phenotypes including; small-eye (Pax6/Sey), dysgenetic lens (Foxe3/dyl), aphakia (Pitx3/ak), eyeless (Pitx3/eyl), and ocular retardation (Vsx2/or-J) [11,41]. In addition, mice homozygous for the eyl allele of Pitx3 exhibit symptoms like Parkinson’s disease in humans [43].

Cataract has been associated with several relatively mild systemic defects that manifest after standard blood or urine laboratory-tests, and serve to highlight the sensitivity of the lens to certain metabolic stresses. Autosomal recessive galactokinase-deficiency cataract results from mutations in the gene coding for galactokinase-1 (GALK1, 17q), the first enzyme in galactose metabolism [44]. GALK1 mutations affect the enzyme coding region resulting in decreased red blood cell galactokinase activity. Autosomal dominant hyperferritinemia-cataract syndrome results from non-coding mutations in the gene for ferritin light chain (FTL, 19q), an iron-storage protein [45]. Specifically, these non-coding FTL mutations are, confined to the iron response element (IRE) located upstream of the coding region, and they result in increased serum ferritin levels. Mutations in the gene for glucosaminyl (N-acetyl) transferase-2 (GCNT2, 6p), a blood-group glycosylation enzyme, underlie autosomal recessive cataract associated with the adult i blood-group phenotype particularly in Japanese and Taiwanese populations [46,47]. Finally, mutations in the gene for solute carrier family-16A member-12 (SLC16A12, 10q), a mono-carboxylic acid transporter, underlie autosomal dominant juvenile cataract plus microcornea and renal glucosuria [48]. In mice, no mutations in the IRE sequence of Ftl have been reported. Mice null for Gcnt2 do not develop early-onset cataract, and mice null for Galk1 do not develop cataract unless they are also transgenic for human aldose reductase [49,50].

Cataract can also present as one aspect of a constellation of relatively severe systemic abnormalities including neurologic disorders (e.g., Cockayne syndrome on 5q and 10q, Walker-Warburg syndrome on 9q and 14q, and Smith-Lemli-Opitz syndrome on 11q). Autosomal dominant forms of systemic cataract include: neurofibromatosis type-2 (NF2, 22q), myotonic dystrophy (DMPK/SIX5, 19q), Marfan syndrome (FBN1, 15q), and brachio-oto-renal-syndrome-1 (EYA1, 8q). Notable autosomal recessive forms of systemic cataract include; inborn errors affecting galactose metabolism (GALT, 9p; GALE,1p) and cholesterol biosynthesis (DHCR7, 11q; CYP27A1, 2q), the premature aging (progeroid) disorders, Werner syndrome (WRN, 8p) and Rothmund Thompson syndrome (RECQL4, 8q), and the spectrum of peroxisomal biogenesis disorders, which include Zellweger syndrome, neonatal adreno-leuko-dystrophy and rhizomelic chondroplasia punctata type-1 (1p, 1q, 6q, 7q, 12p, and 22q).

Finally, the X chromosome alone harbors over 15 syndromic forms of cataract including; the renal disorders - Lowe’s syndrome (OCRL) and Alport syndrome (COL4A5), the lysosomal storage disorder - Fabry’s disease (GLA), and the Nance-Horan cataract-dental syndrome (NHS). Recently, mutations underlying full-blown NHS have been predicted to result in the absence of functional NHS protein (effectively a null), whereas, copy number variations (CNVs) in the NHS gene result in the less severe allelic phenotype of isolated X-linked cataract (CXN) [51]. Mice mutant for Nhs also develop cataract [52].

2.3 Novel genes for Mendelian cataract-- Cat-Map includes at least 16 “orphan” loci for inherited forms of cataract at which the underlying genes remain to be discovered. These include about eleven loci for autosomal dominant cataract (1p, 1q, 2p, 2q, 3q, 14q, 15q, 17p, 17q, 19q, and 20p) [53-65], four loci for autosomal recessive cataract (3p, 7q, 9q, and 19q) [66-70], and one for X-linked cataract (Xq) [71]. In addition, at least 13 genes harboring spontaneous or targeted mutations have been associated with a lens or cataract phenotype in mice but not yet in humans (Ankb, Bin3, Dock5, Dnase2b, Efna5, Gjf1, Gpr161, Gpx1, Hip1, Nrcam, Prox1, Six5, and Sparc) [72-84]. Interestingly, none of these mouse genes are syntenic with the orphan cataract loci in humans. Genotype-phenotype discrepancies between mice and humans are likely to be found. For instance, mice lacking Six5 develop cataract [82,83], whereas, mutations in human SIX5 are associated with branchio-oto-renal syndrome type-2 [85]. Nevertheless, the combined list of orphan loci and mouse mutants suggests that a substantial number of genes for Mendelian forms of cataract remain to be discovered.

3. Genes for age-related cataract

Age-related cataract usually presents after the 4th decade and based on slit-lamp examination may be divided into three clinical types referred to as; nuclear cataract, cortical cataract, and posterior sub-capsular cataract. Each can occur in isolation or in combination (mixed cataract), and may progress to total opacification of the lens.

Genetic epidemiological studies of affected twins and siblings predict that genetic risk factors may account for 14%–48% of the heritability for nuclear cataract, and 24%–75% of the heritability for cortical cataract [86-91]. Overall, age-related cataract is less phenotypically variable than Mendelian forms of cataract, however, the genetic complexity of the former remains largely unknown.

Intuitively, genes underlying Mendelian forms of cataract represent plausible candidates for genetic determinants of age-related cataract [92]. So far, variations in at least eight genes linked with inherited cataract have been associated with age-related cataract. These include EPHA2 (1p), GJA8 (1q), GALT (9p), SLC16A12 (10q), HSF4 (16q), GALK1 (17q), FTL (19q), and CRYAA (21q) [27,29,93-99]. Notably however, the triplet nucleotide (CTG)n repeat expansions underlying adult-onset cataract associated with myotonic dystrophy do not represent a significant risk factor for age-related cataract in the general population [100].

Finally, variations in at least ten other genes not directly associated with inherited cataract have been tentatively implicated in age-related cataract. These include genes that function in antioxidant metabolism (GSTM1, 1p; GSTT1, 22q) [101,102], xenobiotic detoxification (NAT2, 8p) [103], DNA repair (ERCC2, 19q) [104], folate metabolism (MTHFR, 1p) [105], lactose metabolism (LCT, 2q) [106], RNA demethylation (FTO, 16q) [107], lipid/cholesterol transport (APOE4, 19q) [108], kinesin/microtubule motor transport (KLC1, 14q) [109], and one of unknown identity (ARCC1, 6cen) [110].

4. Summary and outlook

Currently, Cat-Map totals almost 200 genes and loci for Mendelian and age-related forms of human cataract spread across all 22 autosomes and the X-chromosome. At least 35 independent loci, including over 20 known genes, have been identified for non-syndromic cataract segregating most often as an autosomal dominant trait with high penetrance in over 190 families worldwide. As much as 70% of autosomal dominant cataract may be accounted for by missense coding mutations in the genes for crystallins, particularly CRYAA, CRYBB2, and CRYGD, and connexins (GJA3, GJA8). Cat-Map also includes over 130 genes and loci for syndromic forms of cataract, many associated with neurologic abnormalities, and over 70 mouse mutants with cataract, some of which point to novel genes for human cataract. Finally, variations in at least eight genes underlying genetic forms of cataract (EPHA2, GJA8, GALT, SLC16A12, HSF4, GALK1, FTL, and CRYAA), and at least 10 other diverse genes have been associated to varying degrees with age-related cataract. Collectively, however, the currently implicated genes are likely to account for a relatively small proportion of the genetic risk for age-related cataract.

The extensive clinical and genetic heterogeneity of Mendelian and age-related forms of cataract limits efforts to make informative genotype-phenotype correlations. Mutations in the same gene may result in different phenotypes. For example, CRYAA mutations can result in cataract +/− microcornea, whereas, CRYAB mutations can result in cataract and/or myopathy. Similarly, mutations or variations in EPHA2, HSF4, and CRYAA are each associated with autosomal dominant, autosomal recessive and age-related forms of cataract. By contrast, mutations in different genes can result in similar phenotypes. For example, mutations in multiple genes have been associated with cataract plus microcornea (e.g., MAF, CRYAA, and GJA8), posterior polar cataract (e.g., PITX3, EPHA2, CHMP4B, and NF2), or sutural cataract (e.g., BFSP2, CRYAB, and NHS). Further, an increasing number of genes have been associated with age-related cataract especially cortical or mixed types (e.g., EPHA2, HSF4, and CRYAA). It is likely that the clinical and genetic heterogeneity of cataract will continue to expand particularly in cases where a non-lens phenotype may present long after the diagnosis of early-onset cataract. Recently, variations in CRYAB and PITX3 have been associated with increased susceptibility to multiple sclerosis and Parkinson’s disease, respectively [111,112], raising awareness about the possibility of acquired neurodegenerative conditions in certain patients with early-onset cataract. Such observations support the notion that a gene-based system will enable a more informative clinical classification of the cataract spectrum disorders.

In the future, family-based linkage studies and case-control association studies in different populations will continue to identify, test and validate genetic determinants of inherited and age-related forms of cataract. Moreover, the advent of next-generation sequencing techniques capable of rapidly deciphering genomic variation in large numbers of individuals will provide powerful insights regarding the molecular genetic basis of cataract; including gene-gene and gene-environment interactions. Ultimately, a comprehensive understanding of the genomic determinants of cataract, coupled with improved phenotyping of animal models, will not only enhance understanding of the molecular biology of lens development and aging but also may translate into non-surgical treatments for cataract, or even lifestyle interventions (e.g., diet) that help to prevent cataract.

In summary, Cat-Map provides a curated portal to access peer-reviewed literature (PubMed) and bioinformatics (EntrezGene) focused on the genetic causes of cataract in humans and mice, and a gene-centric basis to aid clinical classification of cataract.

Acknowledgments

We thank Frank Schottler for website assistance and an anonymous reviewer
for insightful comments. This work was supported by NIH/NEI grants EY012284 (to A.S.) and EY02687 (Core grant for vision research), and by an unrestricted grant to the Department of Ophthalmology and Visual Sciences from Research to Prevent Blindness.

References

  1. Congdon NG, Friedman DS, Lietman T. Important causes of visual impairment in the world today. JAMA. 2003; 290:2057-60. [PMID: 14559961]
  2. Congdon N, Vingerling JR, Klein BE, West S, Friedman DS, Kempen J, O'Colmain B, Wu SY, Taylor HR. Prevalence of cataract and pseudophakia/aphakia among adults in the United States. Arch Ophthalmol. 2004; 122:487-94. [PMID: 15078665]
  3. McCarty CA, Taylor HR. The genetics of cataract. Invest Ophthalmol Vis Sci. 2001; 42:1677-8. [PMID: 11431427]
  4. Hejtmancik JF. Congenital cataracts and their molecular genetics. Semin Cell Dev Biol. 2008; 19:134-49. [PMID: 18035564]
  5. Shiels A, Hejtmancik JF. Genetic origins of cataract. Arch Ophthalmol. 2007; 125:165-73. [PMID: 17296892]
  6. Amaya L, Taylor D, Russell-Eggitt I, Nischal KK, Lengyel D. The morphology and natural history of childhood cataracts. Surv Ophthalmol. 2003; 48:125-44. [PMID: 12686301]
  7. Reddy MA, Francis PJ, Berry V, Bhattacharya SS, Moore AT. Molecular genetic basis of inherited cataract and associated phenotypes. Surv Ophthalmol. 2004; 49:300-15. [PMID: 15110667]
  8. Sperduto RD, Clemons TE, Lindblad AS, Ferris FL, 3rd. Cataract classification using serial examinations in the age-related eye disease study: age-related eye disease study report no. 24. Am J Ophthalmol. 2008; 145:504-8. [PMID: 18201681]
  9. Renwick JH. Eyes on chromosomes. J Med Genet. 1970; 7:239-43. [PMID: 5489092]
  10. Feingold J, Raoul O, See G, Delthil S, Crouzet J, Demailly ML, Morel J. Congenital cataract linked to the Y chromosome. J Genet Hum. 1979; 27:67-9. [PMID: 479855]
  11. Graw J. Mouse models of cataract. J Genet. 2009; 88:469-86. [PMID: 20090208]
  12. Sayers EW, Barrett T, Benson DA, Bolton E, Bryant SH, Canese K, Chetvernin V, Church DM, Dicuccio M, Federhen S, Feolo M, Geer LY, Helmberg W, Kapustin Y, Landsman D, Lipman DJ, Lu Z, Madden TL, Madej T, Maglott DR, Marchler-Bauer A, Miller V, Mizrachi I, Ostell J, Panchenko A, Pruitt KD, Schuler GD, Sequeira E, Sherry ST, Shumway M, Sirotkin K, Slotta D, Souvorov A, Starchenko G, Tatusova TA, Wagner L, Wang Y, John Wilbur W, Yaschenko E, Ye J. Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 2010; 38:D5-16. [PMID: 19910364]
  13. den Dunnen JT, Antonarakis SE. Mutation nomenclature extensions and suggestions to describe complex mutations: a discussion. Hum Mutat. 2000; 15:7-12. [PMID: 10612815]
  14. Plotnikova OV, Kondrashov FA, Vlasov PK, Grigorenko AP, Ginter EK, Rogaev EI. Conversion and compensatory evolution of the gamma-crystallin genes and identification of a cataractogenic mutation that reverses the sequence of the human CRYGD gene to an ancestral state. Am J Hum Genet. 2007; 81:32-43. [PMID: 17564961]
  15. Vanita Reis A, Jung M, Singh D, Sperling K, Singh JR, Burger J. A unique form of autosomal dominant cataract explained by gene conversion between beta-crystallin B2 and its pseudogene. J Med Genet. 2001; 38:392-6. [PMID: 11424921]
  16. Bodker FS, Lavery MA, Mitchell TN, Lovrien EW, Maumenee IH. Microphthalmos in the presumed homozygous offspring of a first cousin marriage and linkage analysis of a locus in a family with autosomal dominant cerulean congenital cataracts. Am J Med Genet. 1990; 37:54-9. [PMID: 2240043]
  17. Hsu CD, Kymes S, Petrash JM. A transgenic mouse model for human autosomal dominant cataract. Invest Ophthalmol Vis Sci. 2006; 47:2036-44. [PMID: 16639013]
  18. Xi JH, Bai F, Gross J, Townsend RR, Menko AS, Andley UP. Mechanism of small heat shock protein function in vivo: a knock-in mouse model demonstrates that the R49C mutation in alpha A-crystallin enhances protein insolubility and cell death. J Biol Chem. 2008; 283:5801-14. [PMID: 18056999]
  19. Shiels A, Mackay D, Ionides A, Berry V, Moore A, Bhattacharya S. A missense mutation in the human connexin50 gene (GJA8) underlies autosomal dominant “zonular pulverulent” cataract, on chromosome 1q. Am J Hum Genet. 1998; 62:526-32. [PMID: 9497259]
  20. Hansen L, Yao W, Eiberg H, Funding M, Riise R, Kjaer KW, Hejtmancik JF, Rosenberg T. The congenital “ant-egg” cataract phenotype is caused by a missense mutation in connexin46. Mol Vis. 2006; 12:1033-9. [PMID: 16971895]
  21. Gong X, Li E, Klier G, Huang Q, Wu Y, Lei H, Kumar NM, Horwitz J, Gilula NB. Disruption of alpha3 connexin gene leads to proteolysis and cataractogenesis in mice. Cell. 1997; 91:833-43. [PMID: 9413992]
  22. White TW, Goodenough DA, Paul DL. Targeted ablation of connexin50 in mice results in microphthalmia and zonular pulverulent cataracts. J Cell Biol. 1998; 143:815-25. [PMID: 9813099]
  23. Shiels A, Bassnett S, Varadaraj K, Mathias R, Al-Ghoul K, Kuszak J, Donoviel D, Lilleberg S, Friedrich G, Zambrowicz B. Optical dysfunction of the crystalline lens in aquaporin-0-deficient mice. Physiol Genomics. 2001; 7:179-86. [PMID: 11773604]
  24. Shiels A, King JM, Mackay DS, Bassnett S. Refractive defects and cataracts in mice lacking lens intrinsic membrane protein-2. Invest Ophthalmol Vis Sci. 2007; 48:500-8. [PMID: 17251442]
  25. Jamieson RV, Farrar N, Stewart K, Perveen R, Mihelec M, Carette M, Grigg JR, McAvoy JW, Lovicu FJ, Tam PP, Scambler P, Lloyd IC, Donnai D, Black GC. Characterization of a familial t(16;22) balanced translocation associated with congenital cataract leads to identification of a novel gene, TMEM114, expressed in the lens and disrupted by the translocation. Hum Mutat. 2007; 28:968-77. [PMID: 17492639]
  26. Shiels A, Bennett TM, Knopf HL, Yamada K, Yoshiura K, Niikawa N, Shim S, Hanson PI. CHMP4B, a novel gene for autosomal dominant cataracts linked to chromosome 20q. Am J Hum Genet. 2007; 81:596-606. [PMID: 17701905]
  27. Shiels A, Bennett TM, Knopf HL, Maraini G, Li A, Jiao X, Hejtmancik JF. The EPHA2 gene is associated with cataracts linked to chromosome 1p. Mol Vis. 2008; 14:2042-55. [PMID: 19005574]
  28. Zhang T, Hua R, Xiao W, Burdon KP, Bhattacharya SS, Craig JE, Shang D, Zhao X, Mackey DA, Moore AT, Luo Y, Zhang J, Zhang X. Mutations of the EPHA2 receptor tyrosine kinase gene cause autosomal dominant congenital cataract. Hum Mutat. 2009; 30:E603-11. [PMID: 19306328]
  29. Jun G, Guo H, Klein BE, Klein R, Wang JJ, Mitchell P, Miao H, Lee KE, Joshi T, Buck M, Chugha P, Bardenstein D, Klein AP, Bailey-Wilson JE, Gong X, Spector TD, Andrew T, Hammond CJ, Elston RC, Iyengar SK, Wang B. EPHA2 is associated with age-related cortical cataract in mice and humans. PLoS Genet. 2009; 5:e1000584 [PMID: 19649315]
  30. Kaul H, Riazuddin SA, Shahid M, Kousar S, Butt NH, Zafar AU, Khan SN, Husnain T, Akram J, Hejtmancik JF, Riazuddin S. Autosomal recessive congenital cataract linked to EPHA2 in a consanguineous Pakistani family. Mol Vis. 2010; 16:511-7. [PMID: 20361013]
  31. Lee JA, Beigneux A, Ahmad ST, Young SG, Gao FB. ESCRT-III dysfunction causes autophagosome accumulation and neurodegeneration. Curr Biol. 2007; 17:1561-7. [PMID: 17683935]
  32. Bornheim R, Muller M, Reuter U, Herrmann H, Bussow H, Magin TM. A dominant vimentin mutant upregulates Hsp70 and the activity of the ubiquitin-proteasome system, and causes posterior cataracts in transgenic mice. J Cell Sci. 2008; 121:3737-46. [PMID: 18940912]
  33. Müller M, Bhattacharya SS, Moore T, Prescott Q, Wedig T, Herrmann H, Magin TM. Dominant cataract formation in association with a vimentin assembly disrupting mutation. Hum Mol Genet. 2009; 18:1052-7. [PMID: 19126778]
  34. Colucci-Guyon E, Portier MM, Dunia I, Paulin D, Pournin S, Babinet C. Mice lacking vimentin develop and reproduce without an obvious phenotype. Cell. 1994; 79:679-94. [PMID: 7954832]
  35. Alizadeh A, Clark JI, Seeberger T, Hess J, Blankenship T, Spicer A, FitzGerald PG. Targeted genomic deletion of the lens-specific intermediate filament protein CP49. Invest Ophthalmol Vis Sci. 2002; 43:3722-7. [PMID: 12454043]
  36. Alizadeh A, Clark J, Seeberger T, Hess J, Blankenship T, FitzGerald PG. Targeted deletion of the lens fiber cell-specific intermediate filament protein filensin. Invest Ophthalmol Vis Sci. 2003; 44:5252-8. [PMID: 14638724]
  37. Alizadeh A, Clark J, Seeberger T, Hess J, Blankenship T, FitzGerald PG. Characterization of a mutation in the lens-specific CP49 in the 129 strain of mouse. Invest Ophthalmol Vis Sci. 2004; 45:884-91. [PMID: 14985306]
  38. Simirskii VN, Lee RS, Wawrousek EF, Duncan MK. Inbred FVB/N mice are mutant at the cp49/Bfsp2 locus and lack beaded filament proteins in the lens. Invest Ophthalmol Vis Sci. 2006; 47:4931-4. [PMID: 17065509]
  39. Bu L, Jin Y, Shi Y, Chu R, Ban A, Eiberg H, Andres L, Jiang H, Zheng G, Qian M, Cui B, Xia Y, Liu J, Hu L, Zhao G, Hayden MR, Kong X. Mutant DNA-binding domain of HSF4 is associated with autosomal dominant lamellar and Marner cataract. Nat Genet. 2002; 31:276-8. [PMID: 12089525]
  40. Lin HJ, Huang YC, Lin JM, Wu JY, Chen LA, Lin CJ, Tsui YP, Chen CP, Tsai FJ. Single-nucleotide polymorphisms in chromosome 3p14.1- 3p14.2 are associated with susceptibility of Type 2 diabetes with cataract. Mol Vis. 2010; 16:1206-14. [PMID: 20664687]
  41. Gould DB, John SW. Anterior segment dysgenesis and the developmental glaucomas are complex traits. Hum Mol Genet. 2002; 11:1185-93. [PMID: 12015278]
  42. Bidinost C, Matsumoto M, Chung D, Salem N, Zhang K, Stockton DW, Khoury A, Megarbane A, Bejjani BA, Traboulsi EI. Heterozygous and homozygous mutations in PITX3 in a large Lebanese family with posterior polar cataracts and neurodevelopmental abnormalities. Invest Ophthalmol Vis Sci. 2006; 47:1274-80. [PMID: 16565358]
  43. Rosemann M, Ivashkevich A, Favor J, Dalke C, Holter SM, Becker L, Racz I, Bolle I, Klempt M, Rathkolb B, Kalaydjiev S, Adler T, Aguilar A, Hans W, Horsch M, Rozman J, Calzada-Wack J, Kunder S, Naton B, Gailus-Durner V, Fuchs H, Schulz H, Beckers J, Busch DH, Burbach JP, Smidt MP, Quintanilla-Martinez L, Esposito I, Klopstock T, Klingenspor M, Ollert M, Wolf E, Wurst W, Zimmer A, de Angelis MH, Atkinson M, Heinzmann U, Graw J. Microphthalmia, parkinsonism, and enhanced nociception in Pitx3 (416insG) mice. Mamm Genome. 2010; 21:13-27. [PMID: 20033184]
  44. Bosch AM, Bakker HD, van Gennip AH, van Kempen JV, Wanders RJ, Wijburg FA. Clinical features of galactokinase deficiency: a review of the literature. J Inherit Metab Dis. 2002; 25:629-34. [PMID: 12705493]
  45. Lachlan KL, Temple IK, Mumford AD. Clinical features and molecular analysis of seven British kindreds with hereditary hyperferritinaemia cataract syndrome. Eur J Hum Genet. 2004; 12:790-6. [PMID: 15280904]
  46. Yu LC, Twu YC, Chang CY, Lin M. Molecular basis of the adult i phenotype and the gene responsible for the expression of the human blood group I antigen. Blood. 2001; 98:3840-5. [PMID: 11739194]
  47. Pras E, Raz J, Yahalom V, Frydman M, Garzozi HJ, Pras E, Hejtmancik JF. A nonsense mutation in the glucosaminyl (N-acetyl) transferase 2 gene (GCNT2): association with autosomal recessive congenital cataracts. Invest Ophthalmol Vis Sci. 2004; 45:1940-5. [PMID: 15161861]
  48. Kloeckener-Gruissem B, Vandekerckhove K, Nurnberg G, Neidhardt J, Zeitz C, Nurnberg P, Schipper I, Berger W. Mutation of solute carrier SLC16A12 associates with a syndrome combining juvenile cataract with microcornea and renal glucosuria. Am J Hum Genet. 2008; 82:772-9. [PMID: 18304496]
  49. Chen GY, Muramatsu H, Kondo M, Kurosawa N, Miyake Y, Takeda N, Muramatsu T. Abnormalities caused by carbohydrate alterations in Ibeta6-N-acetylglucosaminyltransferase-deficient mice. Mol Cell Biol. 2005; 25:7828-38. [PMID: 16107727]
  50. Ai Y, Zheng Z, O'Brien-Jenkins A, Bernard DJ, Wynshaw-Boris T, Ning C, Reynolds R, Segal S, Huang K, Stambolian D. A mouse model of galactose-induced cataracts. Hum Mol Genet. 2000; 9:1821-7. [PMID: 10915771]
  51. Coccia M, Brooks SP, Webb TR, Christodoulou K, Wozniak IO, Murday V, Balicki M, Yee HA, Wangensteen T, Riise R, Saggar AK, Park SM, Kanuga N, Francis PJ, Maher ER, Moore AT, Russell-Eggitt IM, Hardcastle AJ. X-linked cataract and Nance-Horan syndrome are allelic disorders. Hum Mol Genet. 2009; 18:2643-55. [PMID: 19414485]
  52. Huang KM, Wu J, Duncan MK, Moy C, Dutra A, Favor J, Da T, Stambolian D. Xcat, a novel mouse model for Nance-Horan syndrome inhibits expression of the cytoplasmic-targeted Nhs1 isoform. Hum Mol Genet. 2006; 15:319-27. [PMID: 16357105]
  53. Eiberg H, Lund AM, Warburg M, Rosenberg T. Assignment of congenital cataract Volkmann type (CCV) to chromosome 1p36. Hum Genet. 1995; 96:33-8. [PMID: 7607651]
  54. Wang L, Lin H, Shen Y, Huang S, Gu J, Su H, Qi Y. A new locus for inherited nuclear cataract mapped to the long arm of chromosome 1. Mol Vis. 2007; 13:1357-62. [PMID: 17768382]
  55. Gao L, Qin W, Cui H, Feng G, Liu P, Gao W, Ma L, Li P, He L, Fu S. A novel locus of coralliform cataract mapped to chromosome 2p24-pter. J Hum Genet. 2005; 50:305-10. [PMID: 15933805]
  56. Khaliq S, Hameed A, Ismail M, Anwar K, Mehdi SQ. A novel locus for autosomal dominant nuclear cataract mapped to chromosome 2p12 in a Pakistani family. Invest Ophthalmol Vis Sci. 2002; 43:2083-7. [PMID: 12091400]
  57. Abouzeid H, Meire FM, Osman I, ElShakankiri N, Bolay S, Munier FL, Schorderet DF. A new locus for congenital cataract, microcornea, microphthalmia, and atypical iris coloboma maps to chromosome 2. Ophthalmology. 2009; 116:154-62.e1. [PMID: 12091400]
  58. Liu G, Li Y, Ruan Y, Cao W, Xin L, Qian J, Gu J. A new locus for autosomal dominant congenital coronary cataract in a Chinese family maps to chromosome 3q. Mol Vis. 2010; 16:874-9. [PMID: 20508730]
  59. Pras E, Mahler O, Kumar V, Frydman M, Gefen N, Pras E, Hejtmancik JF. A new locus for autosomal dominant posterior polar cataract in Moroccan Jews maps to chromosome 14q22–23. J Med Genet. 2006; 43:e50 [PMID: 17047090]
  60. Vanita Sarhadi VK, Singh D, Reis A, Rueschendorf F, Becker-Follmann J, Jung M, Sperling K. A novel form of “central pouchlike” cataract, with sutural opacities, maps to chromosome 15q21–22. Am J Hum Genet. 2001; 68:509-14. [PMID: 11133359]
  61. Berry V, Ionides AC, Moore AT, Plant C, Bhattacharya SS, Shiels A. A locus for autosomal dominant anterior polar cataract on chromosome 17p. Hum Mol Genet. 1996; 5:415-9. [PMID: 8852669]
  62. Armitage MM, Kivlin JD, Ferrell RE. A progressive early onset cataract gene maps to human chromosome 17q24. Nat Genet. 1995; 9:37-40. [PMID: 7704021]
  63. Bateman JB, Richter L, Flodman P, Burch D, Brown S, Penrose P, Paul O, Geyer DD, Brooks DG, Spence MA. A new locus for autosomal dominant cataract on chromosome 19: linkage analyses and screening of candidate genes. Invest Ophthalmol Vis Sci. 2006; 47:3441-9. [PMID: 16877414]
  64. Li N, Yang Y, Bu J, Zhao C, Lu S, Zhao J, Yan L, Cui L, Zheng R, Li J, Tang J, Zhao K. An autosomal dominant progressive congenital zonular nuclear cataract linked to chromosome 20p12.2-p11.23. Mol Vis. 2006; 12:1506-10. [PMID: 17167408]
  65. Zhang S, Liu M, Dong JM, Yin K, Wang P, Bu J, Li J, Hao YS, Hao P, Wang QK, Wang L. Identification of a genetic locus for autosomal dominant infantile cataract on chromosome 20p12.1-p11.23 in a Chinese family. Mol Vis. 2008; 14:1893-7. [PMID: 18958302]
  66. Pras E, Pras E, Bakhan T, Levy-Nissenbaum E, Lahat H, Assia EI, Garzozi HJ, Kastner DL, Goldman B, Frydman M. A gene causing autosomal recessive cataract maps to the short arm of chromosome 3. Isr Med Assoc J. 2001; 3:559-62. [PMID: 11519376]
  67. Kaul H, Riazuddin SA, Yasmeen A, Mohsin S, Khan M, Nasir IA, Khan SN, Husnain T, Akram J, Hejtmancik JF, Riazuddin S. A new locus for autosomal recessive congenital cataract identified in a Pakistani family. Mol Vis. 2010; 16:240-5. [PMID: 20161816]
  68. Héon E, Paterson AD, Fraser M, Billingsley G, Priston M, Balmer A, Schorderet DF, Verner A, Hudson TJ, Munier FL. A progressive autosomal recessive cataract locus maps to chromosome 9q13-q22. Am J Hum Genet. 2001; 68:772-7. [PMID: 11179024]
  69. Forshew T, Johnson CA, Khaliq S, Pasha S, Willis C, Abbasi R, Tee L, Smith U, Trembath RC, Mehdi SQ, Moore AT, Maher ER. Locus heterogeneity in autosomal recessive congenital cataracts: linkage to 9q and germline HSF4 mutations. Hum Genet. 2005; 117:452-9. [PMID: 15959809]
  70. Riazuddin SA, Yasmeen A, Zhang Q, Yao W, Sabar MF, Ahmed Z, Riazuddin S, Hejtmancik JF. A new locus for autosomal recessive nuclear cataract mapped to chromosome 19q13 in a Pakistani family. Invest Ophthalmol Vis Sci. 2005; 46:623-6. [PMID: 15671291]
  71. Craig JE, Friend KL, Gecz J, Rattray KM, Troski M, Mackey DA, Burdon KP. A novel locus for X-linked congenital cataract on Xq24. Mol Vis. 2008; 14:721-6. [PMID: 18431456]
  72. Moré MI, Kirsch FP, Rathjen FG. Targeted ablation of NrCAM or ankyrin-B results in disorganized lens fibers leading to cataract formation. J Cell Biol. 2001; 154:187-96. [PMID: 11449000]
  73. Ramalingam A, Duhadaway JB, Sutanto-Ward E, Wang Y, Dinchuk J, Huang M, Donover PS, Boulden J, McNally LM, Soler AP, Muller AJ, Duncan MK, Prendergast GC. Bin3 deletion causes cataracts and increased susceptibility to lymphoma during aging. Cancer Res. 2008; 68:1683-90. [PMID: 18339847]
  74. Omi N, Kiyokawa E, Matsuda M, Kinoshita K, Yamada S, Yamada K, Matsushima Y, Wang Y, Kawai J, Suzuki M, Hayashizaki Y, Hiai H. Mutation of Dock5, a member of the guanine exchange factor Dock180 superfamily, in the rupture of lens cataract mouse. Exp Eye Res. 2008; 86:828-34. [PMID: 18396277]
  75. Nishimoto S, Kawane K, Watanabe-Fukunaga R, Fukuyama H, Ohsawa Y, Uchiyama Y, Hashida N, Ohguro N, Tano Y, Morimoto T, Fukuda Y, Nagata S. Nuclear cataract caused by a lack of DNA degradation in the mouse eye lens. Nature. 2003; 424:1071-4. [PMID: 12944971]
  76. Cooper MA, Son AI, Komlos D, Sun Y, Kleiman NJ, Zhou R. Loss of ephrin-A5 function disrupts lens fiber cell packing and leads to cataract. Proc Natl Acad Sci USA. 2008; 105:16620-5. [PMID: 18948590]
  77. Puk O, Loster J, Dalke C, Soewarto D, Fuchs H, Budde B, Nurnberg P, Wolf E, de Angelis MH, Graw J. Mutation in a novel connexin-like gene (Gjf1) in the mouse affects early lens development and causes a variable small-eye phenotype. Invest Ophthalmol Vis Sci. 2008; 49:1525-32. [PMID: 18385072]
  78. Matteson PG, Desai J, Korstanje R, Lazar G, Borsuk TE, Rollins J, Kadambi S, Joseph J, Rahman T, Wink J, Benayed R, Paigen B, Millonig JH. The orphan G protein-coupled receptor, Gpr161, encodes the vacuolated lens locus and controls neurulation and lens development. Proc Natl Acad Sci USA. 2008; 105:2088-93. [PMID: 18250320]
  79. Reddy VN, Giblin FJ, Lin LR, Dang L, Unakar NJ, Musch DC, Boyle DL, Takemoto LJ, Ho YS, Knoernschild T, Juenemann A, Lutjen-Drecoll E. Glutathione peroxidase-1 deficiency leads to increased nuclear light scattering, membrane damage, and cataract formation in gene-knockout mice. Invest Ophthalmol Vis Sci. 2001; 42:3247-55. [PMID: 11726630]
  80. Oravecz-Wilson KI, Kiel MJ, Li L, Rao DS, Saint-Dic D, Kumar PD, Provot MM, Hankenson KD, Reddy VN, Lieberman AP, Morrison SJ, Ross TS. Huntingtin Interacting Protein 1 mutations lead to abnormal hematopoiesis, spinal defects and cataracts. Hum Mol Genet. 2004; 13:851-67. [PMID: 14998932]
  81. Wigle JT, Chowdhury K, Gruss P, Oliver G. Prox1 function is crucial for mouse lens-fibre elongation. Nat Genet. 1999; 21:318-22. [PMID: 10080188]
  82. Klesert TR, Cho DH, Clark JI, Maylie J, Adelman J, Snider L, Yuen EC, Soriano P, Tapscott SJ. Mice deficient in Six5 develop cataracts: implications for myotonic dystrophy. Nat Genet. 2000; 25:105-9. [PMID: 10802667]
  83. Sarkar PS, Appukuttan B, Han J, Ito Y, Ai C, Tsai W, Chai Y, Stout JT, Reddy S. Heterozygous loss of Six5 in mice is sufficient to cause ocular cataracts. Nat Genet. 2000; 25:110-4. [PMID: 10802668]
  84. Gilmour DT, Lyon GJ, Carlton MB, Sanes JR, Cunningham JM, Anderson JR, Hogan BL, Evans MJ, Colledge WH. Mice deficient for the secreted glycoprotein SPARC/osteonectin/BM40 develop normally but show severe age-onset cataract formation and disruption of the lens. EMBO J. 1998; 17:1860-70. [PMID: 9524110]
  85. Hoskins BE, Cramer CH, Silvius D, Zou D, Raymond RM, Orten DJ, Kimberling WJ, Smith RJ, Weil D, Petit C, Otto EA, Xu PX, Hildebrandt F. Transcription factor SIX5 is mutated in patients with branchio-oto-renal syndrome. Am J Hum Genet. 2007; 80:800-4. [PMID: 17357085]
  86. Heiba IM, Elston RC, Klein BE, Klein R. Genetic etiology of nuclear cataract: evidence for a major gene. Am J Med Genet. 1993; 47:1208-14. [PMID: 8291558]
  87. Heiba IM, Elston RC, Klein BE, Klein R. Evidence for a major gene for cortical cataract. Invest Ophthalmol Vis Sci. 1995; 36:227-35. [PMID: 7822150]
  88. Hammond CJ, Snieder H, Spector TD, Gilbert CE. Genetic and environmental factors in age-related nuclear cataracts in monozygotic and dizygotic twins. N Engl J Med. 2000; 342:1786-90. [PMID: 10853001]
  89. Hammond CJ, Duncan DD, Snieder H, de Lange M, West SK, Spector TD, Gilbert CE. The heritability of age-related cortical cataract: the twin eye study. Invest Ophthalmol Vis Sci. 2001; 42:601-5. [PMID: 11222516]
  90. Congdon N, Broman KW, Lai H, Munoz B, Bowie H, Gilber D, Wojciechowski R, Alston C, West SK. Nuclear cataract shows significant familial aggregation in an older population after adjustment for possible shared environmental factors. Invest Ophthalmol Vis Sci. 2004; 45:2182-6. [PMID: 15223793]
  91. Congdon N, Broman KW, Lai H, Munoz B, Bowie H, Gilbert D, Wojciechowski R, West SK. Cortical, but not posterior subcapsular, cataract shows significant familial aggregation in an older population after adjustment for possible shared environmental factors. Ophthalmology. 2005; 112:73-7. [PMID: 15629823]
  92. Moore AT. Understanding the molecular genetics of congenital cataract may have wider implications for age related cataract. Br J Ophthalmol. 2004; 88:2-3. [PMID: 14693758]
  93. Liu Y, Ke M, Yan M, Guo S, Mothobi ME, Chen Q, Zheng F. Association between gap junction protein-alpha 8 polymorphisms and age-related cataract. Mol Biol Rep. 2010; [PMID: 20582632]
  94. Karas N, Gobec L, Pfeifer V, Mlinar B, Battelino T, Lukac-Bajalo J. Mutations in galactose-1-phosphate uridyltransferase gene in patients with idiopathic presenile cataract. J Inherit Metab Dis. 2003; 26:699-704. [PMID: 14707519]
  95. Zuercher J, Neidhardt J, Magyar I, Labs S, Moore AT, Tanner F, Waseem NH, Schorderet D, Munier FL, Bhattacharya SS, Berger W, Kloeckener-Gruissem B. Alterations of the 5′untranslated leader region of SLC16A12 lead to age-related cataract. Invest Ophthalmol Vis Sci. 2010; 51:3354-61. [PMID: 20181839]
  96. Shi Y, Shi X, Jin Y, Miao A, Bu L, He J, Jiang H, Lu Y, Kong X, Hu L. Mutation screening of HSF4 in 150 age-related cataract patients. Mol Vis. 2008; 14:1850-5. [PMID: 18941546]
  97. Okano Y, Asada M, Fujimoto A, Ohtake A, Murayama K, Hsiao KJ, Choeh K, Yang Y, Cao Q, Reichardt JK, Niihira S, Imamura T, Yamano T. A genetic factor for age-related cataract: identification and characterization of a novel galactokinase variant, “Osaka,” in Asians. Am J Hum Genet. 2001; 68:1036-42. [PMID: 11231902]
  98. Faniello MC, Di Sanzo M, Quaresima B, Nistico A, Fregola A, Grosso M, Cuda G, Costanzo F. Bilateral cataract in a subject carrying a C to A transition in the L ferritin promoter region. Clin Biochem. 2009; 42:911-4. [PMID: 19254706]
  99. Bhagyalaxmi SG, Srinivas P, Barton KA, Kumar KR, Vidyavathi M, Petrash JM, Bhanuprakash Reddy G, Padma T. A novel mutation (F71L) in alphaA-crystallin with defective chaperone-like function associated with age-related cataract. Biochim Biophys Acta. 2009; 1792:974-81. [PMID: 19595763]
  100. Winchester CL, Ferrier RK, Sermoni A, Clark BJ, Johnson KJ. Characterization of the expression of DMPK and SIX5 in the human eye and implications for pathogenesis in myotonic dystrophy. Hum Mol Genet. 1999; 8:481-92. [PMID: 9949207]
  101. Güven M, Unal M, Sarici A, Ozaydin A, Batar B, Devranoglu K. Glutathione-S-transferase M1 and T1 genetic polymorphisms and the risk of cataract development: a study in the Turkish population. Curr Eye Res. 2007; 32:447-54. [PMID: 17514530]
  102. Zhou J, Hu J, Guan H. The Association between Copy Number Variations in Glutathione S-transferase M1 and T1 and Age-Related Cataract in a Han Chinese Population. Invest Ophthalmol Vis Sci. 2010; 51:3924-8. [PMID: 20335620]
  103. Tamer L, Yilmaz A, Yildirim H, Ayaz L, Ates NA, Karakas S, Oz O, Yildirim O, Atik U. N-acetyltransferase 2 phenotype may be associated with susceptibility to age-related cataract. Curr Eye Res. 2005; 30:835-9. [PMID: 16251120]
  104. Unal M, Guven M, Batar B, Ozaydin A, Sarici A, Devranoglu K. Polymorphisms of DNA repair genes XPD and XRCC1 and risk of cataract development. Exp Eye Res. 2007; 85:328-34. [PMID: 17637462]
  105. Zetterberg M, Tasa G, Prince JA, Palmer M, Juronen E, Veromann S, Teesalu P, Karlsson JO, Blennow K, Zetterberg H. Methylenetetrahydrofolate reductase genetic polymorphisms in patients with cataract. Am J Ophthalmol. 2005; 140:932-4. [PMID: 16310481]
  106. Karas-Kuzelicki N, Pfeifer V, Lukac-Bajalo J. Synergistic effect of high lactase activity genotype and galactose-1-phosphate uridyl transferase (GALT) mutations on idiopathic presenile cataract formation. Clin Biochem. 2008; 41:869-74. [PMID: 18454942]
  107. Lim LS, Tai ES, Aung T, Tay WT, Saw SM, Seielstad M, Wong TY. Relation of age-related cataract with obesity and obesity genes in an Asian population. Am J Epidemiol. 2009; 169:1267-74. [PMID: 19329528]
  108. Utheim ØA, Ritland JS, Utheim TP, Espeseth T, Lydersen S, Rootwelt H, Semb SO, Elsås T. Apolipoprotein E genotype and risk for development of cataract and age-related macular degeneration. Acta Ophthalmol. 2008; 86:401-3. [PMID: 18498549]
  109. Andersson ME, Zetterberg M, Tasa G, Seibt-Palmer M, Juronen E, Teesalu P, Andersson ME, Zetterberg M, Tasa G, Seibt-Palmer M, Juronen E, Teesalu P, Blennow K, Zetterberg H. Variability in the kinesin light chain 1 gene may influence risk of age-related cataract. Mol Vis. 2007; 13:993-6. [PMID: 17653041]
  110. Iyengar SK, Klein BE, Klein R, Jun G, Schick JH, Millard C, Liptak R, Russo K, Lee KE, Elston RC. Identification of a major locus for age-related cortical cataract on chromosome 6p12-q12 in the Beaver Dam Eye Study. Proc Natl Acad Sci USA. 2004; 101:14485-90. [PMID: 15452352]
  111. van Veen T, van Winsen L, Crusius JB, Kalkers NF, Barkhof F, Pena AS, Polman CH, Uitdehaag BM. [Alpha]B-crystallin genotype has impact on the multiple sclerosis phenotype. Neurology. 2003; 61:1245-9. [PMID: 14610128]
  112. Bergman O, Hakansson A, Westberg L, Nordenstrom K, Carmine Belin A, Sydow O, Olson L, Holmberg B, Eriksson E, Nissbrandt H. PITX3 polymorphism is associated with early onset Parkinson's disease. Neurobiol Aging. 2010; 31:114-7. [PMID: 18420308]