+ All Categories
Home > Documents > Common and Rare Genetic Risk Factors for...

Common and Rare Genetic Risk Factors for...

Date post: 24-Jun-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
14
Common and Rare Genetic Risk Factors for Glaucoma Ryan Wang and Janey L. Wiggs Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts 02114 Correspondence: [email protected] The characterization of genes responsible for glaucoma isthe critical first step toward the development of gene-based diagnostic and screening tests, which could identify individuals at risk for disease before irreversible optic nerve damage occurs. Early-onset forms of glau- coma affecting children and young adults are typically inherited as Mendelian autosomal dominant or recessive traits whereas glaucoma affecting older adults has complex inheri- tance. In this report, we present a comprehensive overview of the genes and genomic regions contributing to inherited glaucoma. RATIONALE FOR DISCOVERY OF GENES CONTRIBUTING TO GLAUCOMA T he characterization of genes responsible for glaucoma is the critical first step toward the development of gene-based diagnostic and screening tests, which could identify individuals at risk for disease before irreversible optic nerve damage occurs. Current glaucoma therapies are limited to reducing elevated intraocular pres- sure (IOP), a major risk factor for the disease. The discovery of disease-related genes could provide new insights into the underlying mo- lecular mechanisms responsible for glaucoma that could form the basis of novel gene-based therapies, including strategies for neuroprotec- tion. Understanding the role of the protein product in health and disease will also provide new insights into the underlying molecular mechanisms responsible for disease. Glaucoma can affect individuals of all ages. Early-onset forms of glaucoma affecting chil- dren and young adults are typically inherited as Mendelian autosomal dominant or recessive traits whereas glaucoma affecting older adults has complex inheritance. Early-onset forms of glaucoma are rare whereas adult-onset forms are more common. RARE VARIANTS CAUSING EARLY-ONSET GLAUCOMA WITH AUTOSOMAL DOMINANT OR AUTOSOMAL RECESSIVE INHERITANCE Compared with adult-onset glaucoma, early- onset forms of glaucoma are rare with incidence ranging from 1/2500 to 1/20,000, depending on the condition and the population (Stoilov et al. 1997; Beijani et al. 2000; Chakrabarti et al. 2010). Genetic variants causing early-onset Editors: Eric A. Pierce, Richard H. Masland, and Joan W. Miller Additional Perspectives on Retinal Disorders: Genetic Approachesto Diagnosis and Treatment available at www.perspectivesinmedicine.org Copyright # 2014 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 101101/cshperspect.a017244 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 1 www.perspectivesinmedicine.org on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/ Downloaded from
Transcript
Page 1: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

Common and Rare Genetic Risk Factorsfor Glaucoma

Ryan Wang and Janey L. Wiggs

Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary,Boston, Massachusetts 02114

Correspondence: [email protected]

The characterization of genes responsible for glaucoma is the critical first step toward thedevelopment of gene-based diagnostic and screening tests, which could identify individualsat risk for disease before irreversible optic nerve damage occurs. Early-onset forms of glau-coma affecting children and young adults are typically inherited as Mendelian autosomaldominant or recessive traits whereas glaucoma affecting older adults has complex inheri-tance. In this report, we present a comprehensive overview of the genes and genomic regionscontributing to inherited glaucoma.

RATIONALE FOR DISCOVERY OF GENESCONTRIBUTING TO GLAUCOMA

The characterization of genes responsiblefor glaucoma is the critical first step toward

the development of gene-based diagnostic andscreening tests, which could identify individualsat risk for disease before irreversible optic nervedamage occurs. Current glaucoma therapies arelimited to reducing elevated intraocular pres-sure (IOP), a major risk factor for the disease.The discovery of disease-related genes couldprovide new insights into the underlying mo-lecular mechanisms responsible for glaucomathat could form the basis of novel gene-basedtherapies, including strategies for neuroprotec-tion. Understanding the role of the proteinproduct in health and disease will also providenew insights into the underlying molecularmechanisms responsible for disease.

Glaucoma can affect individuals of all ages.Early-onset forms of glaucoma affecting chil-dren and young adults are typically inheritedas Mendelian autosomal dominant or recessivetraits whereas glaucoma affecting older adultshas complex inheritance. Early-onset forms ofglaucoma are rare whereas adult-onset formsare more common.

RARE VARIANTS CAUSING EARLY-ONSETGLAUCOMA WITH AUTOSOMALDOMINANT OR AUTOSOMAL RECESSIVEINHERITANCE

Compared with adult-onset glaucoma, early-onset forms of glaucoma are rare with incidenceranging from 1/2500 to 1/20,000, dependingon the condition and the population (Stoilovet al. 1997; Beijani et al. 2000; Chakrabarti etal. 2010). Genetic variants causing early-onset

Editors: Eric A. Pierce, Richard H. Masland, and Joan W. Miller

Additional Perspectives on Retinal Disorders: Genetic Approaches to Diagnosis and Treatment available at

www.perspectivesinmedicine.org

Copyright # 2014 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 101101/cshperspect.a017244

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

1

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 2: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

glaucoma are highly penetrant, although var-iable expressivity may be observed. The genescurrently known to cause early-onset glaucomaare listed in Table 1. All of these genes wereinitially identified using genetic linkage analy-ses of large, multigenerational families. Thesegenes are responsible for congenital glaucoma(CYP1B1 and LTBP2), developmental glaucoma(PITX2, FOXC1, PAX6, and LMX1B), juvenile-onset primary open angle glaucoma (MYOC),and familial normal-tension glaucoma (OPTNand TBK1).

Congenital Glaucoma

Congenital glaucoma (or infantile glaucoma)typically develops before three years of age(Aponte et al. 2010). The condition is primari-ly inherited as an autosomal recessive trait, al-though autosomal dominant families have beendescribed (Sarfarazi et al. 2000). Genetic link-age studies have identified three loci likely tocontain genes contributing to congenital glau-coma: GLC3A (2p21), GLC3B (1p36), andGLC3C (14q22) (Safarazi et al. 1995; Akarsu etal. 1996; Stoilov and Sarfarazi 2002). Two geneshave been discovered: CYP1B1, encoding cyto-chrome P450 1B1 (GLC3A), and LTBP2 (latenttransforming growth factor binding protein 2)(Ali et al. 2009; Beijani et al. 2000). Mutationsin both CYP1B1 and LTBP2 cause autosomalrecessive disease.

CYP1B1 mutations are the most commoncause of congenital glaucoma worldwide (Stoi-

lov et al. 1997). Disease-causing mutations in-clude missense mutations, nonsense mutations,frameshifts, and large-gene deletions (Stoilovet al. 1997; Sarfarazi 2000; Michels-Rauten-strauss et al. 2001). Although these mutationsare highly penetrant, variable expressivity is awell-known feature of CYP1B1-related disease.Genotype–phenotype studies have suggestedthat mutations causing premature truncationof the protein (frameshifts, deletions, inser-tions, and nonsense mutations) cause more se-vere disease with earlier onset than diseasecaused by missense mutations. The gene prod-uct, cytochrome P450 1B1, metabolizes com-plex molecules such as polycyclic aromatic hy-drocarbons and 17b-estradiol (Tokizane et al.2005; Tsuchiya et al. 2005; Sowers et al. 2006).The role of the protein in congenital glaucomais not clear; however, it has been hypothesizedthat cytochrome P450 1B1 activity is responsi-ble for metabolism of compounds involved inocular development (Choudhary et al. 2008;Choudhary et al. 2009).

LTBP2 (Latent transforming growth factor,TGF, b-binding protein 2) was initially identi-fied as the causative gene in patients with early-onset glaucoma from Pakistan and in patients ofGypsy ethnicity (Ali et al. 2009; Narooie-Nejadet al. 2009). The gene is located 1.3 Mb from theGLC3C locus on 14q22; however, LTBP2 muta-tions have not been identified in the family ini-tially used to define the GLC3C locus, suggest-ing that a second congenital glaucoma gene islocated in this region (Sharafieh et al. 2013).

Table 1. Genes responsible for early-onset glaucoma

Disease

Juvenile open-

angle glaucoma

Anterior segment

dysgenesis Congenital glaucoma

Familial normal-

tension glaucoma

Genes MYOC PITX2, FOXC1, PAX6 CY1B1, LTBP2 OPTN, TBK1Protein

functionExtracellular

matrixDevelopmental

regulation of geneexpression

Cytochrome P450 1B1(CYP1B1); extracellularmatrix (LTBP2)

TNF-a signaling,autophagy

Mutation Endoplasmicreticulum stress

Abnormal oculardevelopment

Abnormal oculardevelopment

Increased rate retinalganglion cellapoptosis

Reference(s) Stone et al. 1997 Gould et al. 1997;Mears et al. 1998;Riise et al. 2001

Stoilov et al. 1997;Ali et al. 2009

Rezaie et al. 2002;Morton et al. 2008

R. Wang and J.L. Wiggs

2 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 3: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

The LTBP2 protein is located in the extracellu-lar matrix, where it associates with micofibrilscontaining fibrillin-1 (Jelodari-Mamaghani etal. 2013). Interestingly, LTBP2 mutations havebeen associated with Weill-Marchesani andWeill-Marchesani-like syndrome, a conditioncharacterized by microspherophakia and ab-normal ocular anterior segment development(Haji-Seyed-Javadi et al. 2012). LTBP2 appearsto be a rare cause of congenital glaucoma inethnically diverse populations, such as in theUnited States (Lim et al. 2012).

Developmental Glaucoma

Abnormal development of the anterior seg-ment can lead to dysgenesis of critical ante-rior segment structures involved in glaucoma,including the trabecular outflow pathways andSchlemn’s canal. Developmental glaucoma maybe syndromic (Axenfeld-Rieger syndrome andaniridia) or limited to only ocular involvement.Some patients with anterior segment dysgenesisnever develop glaucoma, whereas others havevery severe disease evident at birth (Gould andJohn 2002). Four genes coding for proteins in-volved in regulation of gene expression can con-tribute to developmental glaucoma (PITX2,FOXC1, PAX6, and LMX1B). Mutations in thesegenes cause autosomal dominant disease withhigh penetrance but variable expressivity bothwithin and among affected families. The phe-notypic features of disease caused by thesegenes, as well as those caused by CYP1B1 andLTBP2, are similar, making it impossible to es-tablish the genetic diagnosis through clinicalexamination alone (Fig. 1). Genetic testing canestablish a molecular diagnosis as well as clarifythe inheritance information necessary for accu-rate genetic counseling.

PITX2 and LMX1B cause syndromic formsof developmental glaucoma (Rieger syndromeand Nail-Patella syndrome, respectively). Ap-proximately 50% of patients with PITX2 muta-tions develop glaucoma (Reis et al. 2012). Re-cently, mutations involving a genomic regiondistal to PITX2 have been identified as diseasecausing (Volkmann et al. 2011). Patients withLMX1B mutations are affected by Nail-Patella

syndrome, which can involve the eye, causing avariety of glaucoma-related features (Chen et al.1998; Dreyer et al. 1998; Mimiwati et al. 2006).Mutations in PAX6 cause abnormal develop-ment of stem cells in the iris and corneal limbus,resulting in a broad range of clinical features,from agenesis of the iris to corneal opacificationcaused by a deficiency of limbal stem cells (Hin-gorani et al. 2009). FOXC1 was initially iden-tified as a gene responsible for iris hypoplasia(Mears et al. 1998). Subsequent studies haveidentified cardiac abnormalities and possiblyother systemic features in patients with muta-tions in these genes (Gould et al. 1997; Swider-ski et al. 1999; Winnier et al. 1999).

Juvenile-Onset Primary Open-AngleGlaucoma

Patients affected by open-angle glaucoma havedisease despite normal anatomic structures.

Adult-onset primary open-angle glaucoma(POAG) develops after 50 years of age, whereasjuvenile-onset (orearly-onset) open-angle glau-

Figure 1. Anterior segment phenotypes in patientswith developmental glaucoma. Anterior segmentphotographs taken with a camera attached to a slit-lamp are shown for four patients with mutationsin genes associated with early-onset glaucoma. No-table features are peripheral corneal opacification(CYP1B1), irido-corneal adhesions (PITX2) andiris atrophy (FOXC1). In the patients with FOXC1mutations the atrophic iris makes it possible to seethe pupillary sphincter muscle, which is normally notvisible because of overlying iris stroma.

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 3

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 4: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

coma affects children and young adults, withonset typically between the ages of 3 and 20. Ju-venile open-angle glaucoma is inherited asan autosomal dominant trait (Wiggs et al.1998) and is characterized by very high IOP(Wiggs et al. 1995; Johnson et al. 1996). Linkagestudies using large families have identified anumber of juvenile open-angle glaucoma loci:GLC1A (1q24.3-q25.2) (Sheffield et al. 1993),GLC1J (9q22) (Wiggs et al. 2004), GLC1K(20p12) (Wiggs et al. 2004), GLC1M (5q22.1-q32) (Wang et al. 2004; Pang et al. 2006; Fanet al. 2007), and GLC1N (15q22-q24) (Wanget al. 2006). Among these loci only one gene,MYOC, coding for myocilin, has been identi-fied (Kubota et al. 1997; Stone et al. 1997). Mis-sense changes in the protein account for 10% ofthe cases of juvenile open-angle glaucoma andalso 3%–5% of the cases of adult-onset POAG(Wiggs et al. 1998; Fingert et al. 1999). Disease-causing mutations are primarily located in theprotein’s olfactomedin domain (Fig. 2) (Adamet al. 1997; Stone et al. 1997; Fingert et al. 1999).Myocilin is an extracellular protein of unknown

function, and loss of the protein function doesnot result in glaucoma (Wiggs and Vollrath2001; Kim et al. 2001), suggesting that the un-derlying genetic mechanism is likely to be again-of-function or dominant-negative effect.Additionally, a common nonsense variant locat-ed in exon 1 (Fig. 2) is a benign polymorphismand does not cause glaucoma or any other phe-notype (Lam et al. 2000). Disease-causing mu-tations reduce the solubility of the protein, lead-ing to protein misfolding and subsequentprotein aggregation in the endoplasmic reticu-lum (Aroca-Aguilar et al. 2010). The misfoldedprotein response may lead to a loss of trabecularmeshwork cells, resulting in high IOP (Joe et al.2003). Interestingly, limiting protein aggrega-tion using the small molecule phenyl butyrate(PBA) causes reduction in IOP both in vitro andin vivo, possibly pointing to novel gene-basedtherapy for patients with myocilin mutations(Yam et al. 2007; Zode et al. 2011, 2012). Recent-ly, common variants in genes coding for multi-plexin collagens XVand XVIII have been shownto modify the severity of myocilin-related glau-

Lys423Glu

Val426Phe

Tyr437His

IIe477Asn

Asn480Lys

Ser502Pro

Pro370Leu

Gly364Val

Glu323Lys

Gly246Arg

Thr293Lys

GLN368X

Thr377Met

Ala445Val

2011 202 243 244 504Olf

LZ

Arg46X

Figure 2. MYOC mutations causing glaucoma. The gene location of selected MYOC mutations known to causeglaucoma are shown. Exon 1 extends from codon 1–201, exon 2 from codon 202–243, and exon 3 from codon244–504. Exon 1 contains a leucine zipper (LZ) and exon 3 contains the Olfactomedin domain (Olf ). Mutationscausing early-onset glaucoma are shown in blue, adult-onset glaucoma in red, and a nonsense mutation knownto be a benign polymorphism in green.

R. Wang and J.L. Wiggs

4 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 5: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

coma, suggesting that myocilin may interactwith these protein in the extracellular matrixand/or in maintaining the integrity of the tra-becular outflow pathways (Liton et al. 2005;Wiggs et al. 2013b).

Familial Normal-Tension Glaucoma

Normal-tension glaucoma (NTG) is a type ofopen-angle glaucoma characterized by opticnerve degeneration despite normal IOP. MostNTG patients have adult-onset disease; how-ever, rarely, the disease can have early onsetwith autosomal dominant inheritance. Twogenes have been associated with familial NTG:OPTN and TBK1 (Rezaie et al. 2002; Mortonet al. 2008; Fingert et al. 2011).

Rare missense mutations in OPTN, whichencodes optineurin and particularly the E50Kmutation, cause a severe early-onset familialform of NTG. (Aung et al. 2005; Morton et al.2008). Optineurin is involved in basic cellularfunctions including protein trafficking, main-tenance of the Golgi apparatus, as well as theNF-kB pathway, antiviral responses, and anti-bacterial signaling (Ying 2012). Of interest, op-tineurin interacts with Tank1 (TBK1), whichhas also been implicated in familial NTG (Mor-ton et al. 2008). The optineurin E50K mutationcauses the protein to become inactive and/orinsoluble (Minegeshi et al. 2013). This muta-tion also enhances the interaction of optineurinwith Tank1 (Fingert et al. 2011).

COMMON VARIANTS CONTRIBUTINGTO ADULT-ONSET GLAUCOMA WITHCOMPLEX INHERITANCE

POAG, adult-onset NTG, pseudoexfoliationglaucoma, and primary angle-closure glaucomaare common traits with complex inheritance.Genetic variants contributing to these condi-tions have been identified using genome-wideassociation approaches (Thorleifsson et al.2007, 2010; Burdon et al. 2011; Vithana et al.2012; Wiggs et al. 2012, 2013a). Generally, vari-ants contributing to common complex traitssuch as adult-onset glaucoma have small indi-vidual effects on disease development. Aggregat-ed effects from multiple risk factors, includingenvironmental risk factors, can have a larger im-pact. Robust genetic associations have been ob-served for POAG (CDKN2BAS [Burdon et al.2011; Wiggs et al. 2012], SIX1/SIX6 [Wiggset al. 2012], CAV1/CAV2 [Thorliefsson et al.2010; Wiggs et al. 2011], TMCO1 [Burdonet al. 2011]), NTG (CDKN2BAS [Wiggs et al.2012] and a regulatory region on 8q22 [Wiggset al. 2012]), pseudoexfoliation (LOXL1 [Thor-leifsson et al. 2007]), and primary angle-closureglaucoma (PLEKHA7 and COL11A1 [Vithanaet al. 2012]) (Table 2). Additionally, genetic var-iants associated with quantitative ocular traitsthat are risk factors for common forms of glau-coma have also been identified. These includeIOP (van Koolwijk et al. 2012), central cornealthickness (CCT) (Vithana et al. 2011; Lu et al.2013), and optic nerve parameters including

Table 2. Genes/loci contributing to adult-onset glaucoma

Gene Disease Function Reference(s)

CDKN2BAS POAG, NTG Cell cycle regulation, TGF-b signaling Burdon et al. 2011;Wiggs et al. 2012

TMCO1 POAG Unknown Burdon et al. 2011SIX1/SIX6 POAG Optic nerve development Fan et al. 2011; Ramdas

et al. 20118q22

enhancerNTG Regulation of fluid production in

ciliary body and choroid plexusWiggs et al. 2012

CAV1/CAV2 POAG Endothelial membranes Thorleifsson et al. 2010LOXL1 Pseudoexfoliation Elastinogenesis and maintenance Thorleifsson et al. 2007PLEKHA7 Angle-closure glaucoma Adherens junction protein Vithana et al. 2012COL11A1 Angle-closure glaucoma Sclera development Vithana et al. 2012

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 5

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 6: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

cup-to-disc ratio and optic nerve area (Ramdaset al. 2010; Macgregor et al. 2010; Axenovichet al. 2011) (Table 3).

The clinical features of POAG generally in-clude both elevated IOP and optic nerve degen-eration. Patients with optic nerve disease with-out elevation of IOP are referred to as the NTGsubgroup. Large genome-wide association stud-ies (GWAS) have been completed for POAGin the Icelandic population (Thorleifsson etal. 2010), Australian population (Burdon et al.2011), and in the United States (Wiggs et al.2012). The Icelandic study identified a signif-icant association between genetic variants lo-cated in the CAV1/CAV2 genomic region andPOAG. This finding has been replicated in acase-control sample from the US (Wiggs et al.2011) and recently, CAV1/CAV2 variants havebeen shown to be preferentially associated withpatients who have loss of the central visual fieldearly in the disease (Loomis et al. 2014). Both theAustralian and the US studies identified sig-nificant associations with CDKN2BAS, a longnoncoding antisense RNA that inhibits the ex-pression of CDKN2B, an inhibitor of CDK4, aprotein involved in promoting cell division(Burdon et al. 2011; Wiggs et al. 2012). The Aus-tralian study showed significant association withvariants in the TMCO1 genomic region, a genethat also appears to be associated with elevatedIOP (see below). The US study identified sig-nificant association with variants in the SIX1/SIX6 genomic region; these are also associatedwith the cup-to disc ratio, a quantitative opticnerve parameter (Ramdas et al. 2010; Wiggset al. 2012).

NTG

Optic nerve degeneration without IOP eleva-tion (,21 mmHg) is the defining clinical fea-ture of NTG. Identifying the genetic factorsthat contribute to this condition could provideimportant insights into neuroprotective ther-apies for glaucoma. An analysis of the NTGsubgroup in a large genome-wide associationstudy of POAG using a United States case-con-trol sample identified common variants in theCDKN2BAS region and a novel regulatory re-gion on chromosome 8q22 as significant riskfactors (Wiggs et al. 2012). The 8q22 region isa DNaseI hypersensitivity site that has high ac-tivity in the choroid plexus, which is responsiblefor the production of cerebrospinal fluid, andthe ciliary body, which is responsible for theproduction of aqueous humor. Interestingly, re-cent hypotheses suggest that the translaminarpressure across the lamina cribrosa may be arisk factor for optic nerve damage, and this pres-sure gradient is defined by the difference be-tween the IOP and cerebrospinal fluid pressure(Jonas and Wang 2013).

Pseudoexfoliation Syndrome

Pseudoexfoliation syndrome is a generalizeddisorder of extracellular matrix that results ina chronic deposition of fibrillar aggregatesthroughout the ocular anterior segment. Accu-mulation of the fibrillar material in the trabec-ular meshwork is thought to contribute to thedevelopment of glaucoma associated with thiscondition (Schlotzer-Schrehardt et al. 1995;

Table 3. Selected genes/loci contributing to quantitative ocular traits related to glaucoma

Gene Trait Function Reference(s)

CDKN2BAS Cup-to-disc-ratio Cell cyle regulation, TGF-b signaling Fan et al. 2011; Ramdas et al. 2011SIX1/SIX6 Cup-to-disc-ratio Developmental protein Fan et al. 2011ATOH7 Optic nerve area Developmental protein Macgregor et al. 2010TMCO1 IOP Unknown function van Koolwijk et al. 2012GAS7 IOP Unknown function van Koolwijk et al. 2012ZNF469 CCT Regulates expression of extracellular

matrix componentsAbu et al. 2008

COL5A1 CCT Extracellular matrix Lu et al. 2013COL8A2 CCT Extracellular matrix Vithana et al. 2011

R. Wang and J.L. Wiggs

6 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 7: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

Naumann et al. 1998). A genome-wide associ-ation study conducted in the Icelandic popula-tion first identified common variants in theLOXL1 gene as significant risk factors (Thor-leifsson et al. 2007). Subsequently, associationbetween LOXL1 gene variants and pseudoex-foliation has been replicated in populationsworldwide (Fan et al. 2011). LOXL1 (lysyl-oxi-dale like 1) is necessary for formation and main-tenance of elastin. It is not yet known howLOXL1 variants influence development of pseu-doexfoliation; however, recent evidence sug-gests that the variants associated with diseasecause a decrease in gene expression, possiblyresulting in reduced enzyme activity (Schlot-zer-Schrehardt 2009). The LOXL1 risk allelesare very common in pseudoexfoliation cases(95%–99%) but are also common in the nor-mal population (60%–80%) suggesting thatthe gene variants are necessary but not sufficientfor disease development (Fan et al. 2011). Re-cently, residence in northern latitudes has beenidentified as an environmental risk factor forthis condition (Stein et al. 2011; Kang et al.2012).

Primary Angle-Closure Glaucoma

Primary angle-closure glaucoma is commonin Asian countries. A recent genome-wide as-sociation study using five independent Asiancase-control samples identified three genomicregions statistically associated with POAG:rs3753841 (COL11A1), rs1015213 (located be-tween PCMTD1 and ST18), and rs11024102(PLEKHA7) (Vithana et al. 2012). All of thesesingle-nucleotide polymorphisms were alsoreplicated in a combined Australian-Nepalesecase-control sample (Awadalla et al. 2013).COL11A1 is expressed in the sclera, and muta-tions in COL11A1 are known to cause Stickler’ssyndrome, a condition that includes myopia,among other ocular features (Richards et al.1996). As primary angle-closure glaucoma canbe related to hyperopia, the genome-wide as-sociation study results may point to a role forCOL111A1 in development of refractive errors,possibly because of abnormal scleral develop-ment.

Quantitative Ocular Traits that Are RiskFactors for Glaucoma

For traits with complex inheritance, it can beuseful to identify genetic factors influencingthe underlying individual traits, such as IOP,optic nerve parameters, and CCT, that contrib-ute to the overall disease (Charlesworth et al.2010). These traits are quantitative, highly her-itable, and show substantial variation in humanpopulations. IOP is the only modifiable riskfactor for glaucoma. Recent genome-wide anal-yses using normal populations have identifiedtwo genes significantly associated with IOP,GAS7 and TMCO1 (van Koolwijk et al. 2012).Similar analyses for optic nerve parameters as-sociated with glaucoma risk have identifiedCDKN2BAS and SIX1SIX6 as genetic risk fac-tors contributing to cup-to-disc-ratio (Ramdaset al. 2010; Fan et al. 2011) and ATOH7 as animportant determinant of optic nerve size(Macgregor et al. 2010). Populations fromaround the world have been used to identifygenetic factors contributing to CCT, one ofthe most heritable of the quantitative oculartraits (Toh et al. 2005). A recent study fromthe International Glaucoma Genetics Consor-tium (IGGC) identified 16 loci significantly as-sociated with CCT (Fig. 3) (Lu et al. 2013).Pathway analyses suggested that collagen andextracellular matrix pathways are importantregulators of CCT (Lu et al. 2013).

COMPLEX INTERACTIONS: GENE–ENVIRONMENT, GENE–GENEINTERACTIONS, AND EPIGENETICEFFECTS

Complex genetic and gene–environment inter-actions are expected to contribute to the geneticarchitecture of common adult-onset disorders.Nutritional factors, such as dietary fat, antiox-idant intake, and other lifestyle factors includ-ing smoking and postmenopausal hormone usemay influence the development of POAG (Pas-quale and Kang 2009). A gene–environmentinteraction involving hormone replacementtherapy and NOS3 (the gene coding for nitricoxide synthase 3) has been identified as a risk

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 7

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 8: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

factor for POAG (Kang et al. 2010). The estro-gen metabolism pathway is also associated withPOAG in women (Pasquale et al. 2013). Gene–gene interactions contributing to POAG havebeen suggested for WDR36 and p53 (both influ-encing apoptosis) (Blanco-Marchite et al. 2011)and for genetic variants in ATOH7 and SIX1/SIX6 (Fan et al. 2011).

BIOLOGICAL PATHWAYS AND PROCESSESSUGGESTED BY GENETIC VARIANTSCONTRIBUTING TO GLAUCOMA

Genome-wide association studies have identi-fied multiple genes potentially involved in glau-coma pathogenesis; however, the stringent cor-rection for multiple testing and type I erroridentifies only variants with the largest maineffects as statistically significant (Manolio et al.

2008). Many biologically meaningful associa-tions of smaller effect size may go undetected;including variants with smaller effect size in sys-tematic groups could reveal novel biologicalpathways or systems underlying disease sus-ceptibility. Pathway-based analysis groups genevariants into biologically meaningful entities todistinguish the actual from the false-positiveassociations. In short, this type of analysis canbe used to uncoveradditional genotype–pheno-type associations that the single-allele genome-wide association analysis may have missed.Hypothesis-independent pathway analyses sug-gest that the underlying molecular pathogene-sis of POAG involves a broad range of biologi-cal processes, including cell adhesion, immuneresponsiveness, energy metabolism, and neuro-transmission (Cooke Bailey et al. 2013). Identi-fication of genes contributing to both early-on-

DCLK1

Optic nerveparameters

NTGPOAG

IOP

CCT

SKYL1/LTBP3

CDC7/TGFBR3

SIX1/SIX6

CAV1/CAV2

FNDC3B

COL8A2 CHSY1HS3ST3B1/PMP22

ZNF469RXRA/COL5A1

FGF9/SGCGLCN12/PTGDS

CWC27/ADAMTS6TGP1 GPR15 FOXO1LRRK1 USP37 AKAP13

TMCO1

GAS7

8q22CDKN2BAS

SALL1 CHEK2

ATOH7

Figure 3. Gene and genomic regions with common variants that contribute to glaucoma and glaucoma-relatedocular quantitative traits. Variants contributing to both primary open-angle glaucoma and normal tensionglaucoma as well as selected ocular quantitative traits (CCT, IOP, and optic nerve parameters) are shown. Genenames are presented in dark font. Genes contributing to POAG are contained in the orange ellipse, NTG in thepurple ellipse, CCT in the blue ellipse, IOP in the red ellipse, and optic nerve parameters in the green ellipse.Genes that contribute to more than one trait are indicated by their placement in the overlapping regions. Forexample, CDKN2BAS contributes to NTG, POAG, and to optic nerve parameters.

R. Wang and J.L. Wiggs

8 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 9: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

set and adult-onset forms of glaucoma are alsohighlighting biological pathways and processesof importance to glaucoma pathogenesis. Sev-eral of these are discussed below.

Extracellular Matrix Metabolism

Maintenance of extracellular matrix is emergingas an important theme in glaucoma pathogen-esis (Acott et al. 2008; Crawford Downs et al.2011). Several of the genes contributing to ear-ly-onset glaucoma (MYOC, LTBP2, COL18A1)and adult-onset glaucoma (LOXL1) have im-portant roles in extracellular matrix (Ueda etal. 2002; Ali et al. 2009; Sethi et al. 2011; Wiggset al. 2013b), and three important genes relatedto CCT impact extracellular matrix (Lu et al.2013). Further investigation of genes shown tocontribute to extracellular processes in oculartissues relevant to glaucoma could be of interest.

Transforming Growth Factor b (TGF-b)Signaling

TGF-b I and TGF-b II appear to have importantroles in optic nerve degeneration (Zode et al.2009; Fuchshofer 2011) as well as trabecularmeshwork function (Sethi et al. 2011; Fuch-shofer 2012). LTPB2 (associated with congenitalglaucoma and anterior segment dysgenesis) andCDKN2BAS (associated with POAG, NTG) arepart of the overall TGF-b signaling pathway.Additionally, NTG pathway analysis showed sig-nificant association with the TGF-b signalingpathway overall (Wiggs et al. 2012).

Tumor Necrosis Factor a (TNF-a) Signaling

Both genes contributing to familial NTG(OPTN, TBK1) can impact TNF-a signaling(Fingert et al. 2011). Blocking of the TNF-areceptor in an animal model of glaucoma ap-pears to be neuroprotective (Roh et al. 2012).These results suggest that TNF-a signaling canhave an important role in ganglion cell apopto-sis in glaucoma. Higher levels of soluble TNF-ahave been identified in women before the devel-opment of NTG (Kang et al. 2013), a findingconsistent with this hypothesis.

Estrogen Metabolism

The hormone 17b-estradiol can regulate ex-pression of both CAV1 and NOS3 (both associ-ated with POAG), and loss of caveolin-1 func-tion can cause increased expression of NOS3(Zhou et al. 2009; Sud et al. 2010), suggestingan interaction between these proteins. Addi-tionally, P450 1B1 (CYP1B1, associated withcongenital glaucoma) is an important factorin estrogen metabolism (Hanna et al. 2000).Gender effects in glaucoma have also been sug-gested by studies showing that estrogen recep-tors are expressed on the retinal ganglion cells ofthe optic nerve (Munaut et al. 2001), and thatestrogen may have a neuroprotective effect inanimal models of glaucoma (Zhou et al. 2007;Russo et al. 2008). Further studies examininggender effects in POAG, and gender interactionswith genes contributing to this disorder will beof interest.

CONCLUDING REMARKS

Genes responsible for both early-onset andadult-onset glaucoma have been identified us-ing genetic and genomic technologies and ap-proaches. Family-based genetic linkage analyseshave yielded disease-causing genes for early-onset glaucoma, whereas genome-wide asso-ciation studies have identified genes and geno-mic regions contributing to adult-onset formsof the disease. Biological pathways contribut-ing to glaucoma and other complex interac-tions (gene–gene and gene–environment) areemerging. Future directions for new gene dis-covery include whole exome sequencing andother next generation sequencing technologies,genome-wide association studies using largernumbers of cases and controls and includingrare variant analyses, and further evaluation ofthe contributions of biological pathways, gene–gene and gene–environment interactions.

REFERENCES

Abu A, Frydman M, Marek D, Pras E, Nir U, Reznik-Wolf H,Pras E. 2008. Deleterious mutations in the Zinc-Finger469 gene cause brittle cornea syndrome. Am J Hum Genet82: 1217–1222.

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 9

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 10: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

Acott TS, Kelley MJ. 2008. Extracellular matrix in the tra-becular meshwork. Exp Eye Res 86: 543–561.

Adam MF, Belmouden A, Binisti P, Brezin AP, Valtot F, Be-chetoille A, Dascotte JC, Copin B, Gomez L, Chaventre A,et al. 1997. Recurrent mutations in a single exon encodingthe evolutionarily conserved olfactomedin-homologydomain of TIGR in familial open-angle glaucoma.Hum Mol Genet 6.12: 2091–2097.

Akarsu A, Nurten M, Erol Turacli S, Gulderen Aktan, MagdaBarsoum-Homsy, Line Chevrette, B, Sitki Sayli, MansoorSarfarazi. 1996. A second locus (GLC3B) for primarycongenital glaucoma (Buphthalmos) maps to the 1p36region. Hum Mol Genet 5: 1199–1203.

Ali M, McKibbin M, Booth A, Parry DA, Jain P, RiazuddinSA, Hejtmancik JF, Khan SN, Firasat S, Shires M, et al.2009. Null mutations in LTBP2 cause primary congenitalglaucoma. Am J Hum Genet 84: 664–671.

Aponte EP, Diehl N, Mohney BG. 2010. Incidence and clin-ical characteristics of childhood glaucoma: A population-based study. Arch Ophthalmol 128: 478–482.

Aroca-Aguilar JD, Martınez-Redondo F, Sanchez-Sanchez F,Coca-Prados M, Escribano J. 2010. Functional role ofproteolytic processing of recombinant myocilin in self-aggregation. Invest Ophthalmol Vis Sci 51: 72–78.

Aung T, Rezaie T, Okada K, Viswanathan AC, Child AH,Brice G, Bhattacharya SS, Lehmann OJ, Sarfarazi M,Hitchings RA. 2005. Clinical features and course of pa-tients with glaucoma with the E50K mutation in the op-tineurin gene. Invest Ophthalmol Vis Sci 46: 2816–2822.

Awadalla MS, Thapa SS, Hewitt AW, Burdon KP, Craig JE.2013. Association of genetic variants with primary angleclosure glaucoma in two different populations. PLoSONE 8: e67903.

Axenovich T, Zorkoltseva I, Belonogova N, van KoolwijkLM, Borodin P, Kirichenko A, Babenko V, Ramdas WD,Amin N, Despriet DD, et al. 2011. Linkage and associa-tion analyses of glaucoma related traits in a large pedigreefrom a Dutch genetically isolated population. J Med Ge-net 48: 802–809.

Bejjani BA, Stockton DW, Lewis RA, Tomey KF, Dueker DK,Jabak M, Astle WF, Lupski JR. 2000. Multiple CYP1B1mutations and incomplete penetrance in an inbred pop-ulation segregating primary congenital glaucoma suggestfrequent de novo events and a dominant modifier locus.Hum Mol Genet 9: 367–374.

Blanco-Marchite C, Sanchez-Sanchez F, Lopez-Garrido MP,Inigez-de-Onzono M, Lopez-Martınez F, Lopez-SanchezE, Alvarez L, Rodrıguez-Calvo PP, Mendez-Hernandez C,Fernandez-Vega L, et al. 2011. WDR36 and P53 genevariants and susceptibility to primary open-angle glau-coma: Analysis of gene-gene interactions. Invest Ophthal-mol Vis Sci 52: 8467–8478.

Burdon KP, Macgregor S, Hewitt AW, Sharma S, Chidlow G,Mills RA, Danoy P, Casson R, Viswanathan AC, Liu JZ, etal. 2011. Genome-wide association study identifies sus-ceptibility loci for open angle glaucoma at TMCO1 andCDKN2B-AS1. Nat Genet 43: 574–578.

Chakrabarti S, Ghanekar Y, Kaur K, Kaur I, Mandal AK, RaoKN, Parikh RS, Thomas R, Majumder PP. 2010. A poly-morphism in the CYP1B1 promoter is functionally asso-ciated with primary congenital glaucoma. Hum Mol Ge-net 19: 4083–4090.

Charlesworth J, Kramer PL, Dyer T, Diego V, Samples JR,Craig JE, Mackey DA, Hewitt AW, Blangero J, Wirtz MK.2010. The path to open-angle glaucoma gene discovery:Endophenotypic status of intraocular pressure, cup-to-disc ratio, and central corneal thickness. Invest Ophthal-mol Vis Sci 51: 3509–3514.

Chen H, Lun Y, Ovchinnikov D, Kokubo H, Oberg KC,Pepicelli CV, Can L, Lee B, Johnson RL. 1998. Limb andkidney defects in Lmx1b mutant mice suggest an involve-ment of LMX1B in human nail patella syndrome. NatGenet 19: 51–55.

Choudhary D, Jansson I, Sarfarazi M, Schenkman JB. 2008.Characterization of the biochemical and structural phe-notypes of four CYP1B1 mutations observed in individ-uals with primary congenital glaucoma. PharmacogenetGenomics 18: 665–676.

Choudhary D, Jansson I, Schenkman JB. 2009. CYP1B1, adevelopmental gene with a potential role in glaucomatherapy. Xenobiotica 39: 606–615.

Cooke Bailey JN, Sobrin L, Pericak-Vance MA, Haines JL,Hammond CJ, Wiggs JL. 2013. Advances in the genomicsof common eye diseases. Hum Mol Genet 22: R59–R65.

Crawford Downs J, Roberts MD, Sigal IA. 2011. Glaucom-atous cupping of the lamina cribrosa: A review of theevidence for active progressive remodeling as a mecha-nism. Exp Eye Res 93: 133–140.

Dreyer SD, Zhou G, Baldini A, Winterpacht A, Zabel B, ColeW, Johnson RL, Lee B. 1998. Mutations in LMX1B causeabnormal skeletal patterning and renal dysplasia in nailpatella syndrome. Nat Genet 19: 47–50.

Fan BJ, Ko WC, Wang DY, Canlas O, Ritch R, Lam DS, PangCP. 2007. Fine mapping of new glaucoma locus GLC1Mand exclusion of neuregulin 2 as the causative gene. MolVis 13: 779–784.

Fan BJ, Wang DY, Pasquale LR, Haines JL, Wiggs JL. 2011.Genetic variants associated with optic nerve vertical cup-to-disc ratio are risk factors for primary open angle glau-coma in a US Caucasian population. Invest OphthalmolVis Sci 52: 1788–1792.

Fingert JH, Heon E, Liebmann JM, Yamamoto T, Craig JE,Rait J, Kawase K, Hoh ST, Buys YM, Dickinson J, et al.1999. Analysis of myocilin mutations in 1703 glaucomapatients from five different populations. Hum Mol Genet8: 899–905.

Fingert JH, Robin AL, Stone JL, Roos BR, Davis LK, ScheetzTE, Bennett SR, Wassink TH, Kwon YH, Alward WL, etal. 2011. Copy number variations on chromosome 12q14in patients with normal tension glaucoma. Hum MolGenet 20: 2482–2494.

Fuchshofer R. 2011. The pathogenic role of transforminggrowth factor-b2 in glaucomatous damage to the opticnerve head. Exp Eye Res 93: 165–169.

Fuchshofer R, Tamm ER. 2012. The role of TGF-b in thepathogenesis of primary open-angle glaucoma. Cell Tis-sue Res 347: 279–290.

Gould DB, John SW. 2002. Anterior segment dysgenesis andthe developmental glaucomas are complex traits. HumMol Genet 11: 1185–1193.

Gould DB, Mears AJ, Pearce WG, Walter MA. 1997. Auto-somal dominant Axenfeld-Rieger anomaly maps to 6p25.Am J Hum Genet 61: 765–768.

R. Wang and J.L. Wiggs

10 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 11: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

Haji-Seyed-Javadi R, Jelodari-Mamaghani S, Paylakhi SH,Yazdani S, Nilforushan N, Fan JB, Klotzle B, MahmoudiMJ, Ebrahimian MJ, Chelich N, et al. 2012. LTBP2 mu-tations cause Weill-Marchesani and Weill-Marchesani-like syndrome and affect disruptions in the extracellularmatrix. Hum Mutat 33: 1182–1187.

Hanna IH, Dawling S, Roodi N, Guengerich FP, Parl FF.2000. Cytochrome P450 1B1 (CYP1B1) pharmacogenet-ics: Association of polymorphisms with functional dif-ferences in estrogen hydroxylation activity. Cancer Res 60:3440–3444.

Hingorani M, Williamson KA, Moore AT, Van Heyningen V.2009. Detailed ophthalmologic evaluation of 43 individ-uals with PAX6 mutations. Invest Ophthalmol Vis Sci 14:2581–2590.

Jelodari-Mamaghani S, Haji-Seyed-Javadi R, Suri F, Nilfor-ushan N, Yazdani S, Kamyab K, Elahi E. 2013. Contribu-tion of the latent transforming growth factor-b bindingprotein 2 gene to etiology of primary open angle glauco-ma and pseudoexfoliation syndrome. Mol Vis 19: 333–347.

Joe MK, Sohn S, Hur W, Moon Y, Choi YR, Kee C. 2003.Accumulation of mutant myocilins in ER leads to ERstress and potential cytotoxicity in human trabecularmeshwork cells. Biochem Biophys Res Commun 312:592–600.

Johnson AT, Richards JE, Boehnke M, Stringham HM, Her-man SB, Wong DJ, Lichter PR. 1996. Clinical phenotypeof juvenile-onset primary open-angle glaucoma linked tochromosome 1q. Ophthalmology 103: 808–814.

Jonas JB, Wang N. 2013. Cerebrospinal fluid pressure andglaucoma. J Ophthalmic Vis Res 8: 257–263.

Kang JH, Wiggs JL, Rosner BA, Hankinson SE, Abdrabou W,Fan BJ, Haines J, Pasquale LR. 2010. Endothelial nitricoxide synthase gene variants and primary open-angleglaucoma: Interactions with sex and postmenopausalhormone use. Invest Ophthalmol Vis Sci 51: 971–979.

Kang JH, Loomis S, Wiggs JL, Stein JD, Pasquale LR. 2012.Demographic and geographic features of exfoliationglaucoma in 2 United States-based prospective cohorts.Ophthalmology 119: 27–35.

Kang JH, Wiggs JL, Pasquale LR. 2013. A nested case controlstudy of plasma ICAM-1, E-selectin and TNF receptor 2levels, and incident primary open-angle glaucoma. InvestOphthalmol Vis Sci 54: 1797–1804.

Kim BS, Savinova OV, Reedy MV, Martin J, Lun Y, Gan L,Smith RS, Tomarev SI, John SW, Johnson RL. 2001. Tar-geted disruption of the myocilin gene (Myoc) suggeststhat human glaucoma-causing mutations are gain offunction. Mol Cell Biol 21: 7707–7713.

Kubota R, Noda S, Wang Y, Minoshima S, Asakawa S, KudohJ, Mashima Y, Oguchi Y, Shimizu N. 1997. A novel myo-sin-like protein (myocilin) expressed in the connectingcilium of the photoreceptor: Molecular cloning, tissueexpression, and chromosomal mapping. Genomics 41:360–369.

Lam DS, Leung YF, Chua JK, Baum L, Fan DS, Choy KW,Pang CP. 2000. Truncations in the TIGR gene in individ-uals with and without primary open-angle glaucoma.Invest Ophthalmol Vis Sci 41: 1386–1391.

Lim SH, Tran-Viet KN, Yanovitch TL, Freedman SF, KlemmT, Call W, Powell C, Ravichandran A, Metlapally R, Nad-

ing EB, et al. 2013. CYP1B1, MYOC, and LTBP2 muta-tions in primary congenital glaucoma patients in theUnited States. Am J Ophthalmol 155: 508–517.

Liton PB, Liu X, Stamer WD, Challa P, Epstein DL, GonzalezP. 2005. Specific targeting of gene expression to a subsetof human trabecular meshwork cells using the chitinase3-like 1 promoter. Invest Ophthalmol Vis Sci 46: 183–190.

Loomis SJ, Kang JH, Weinreb RN, Yaspan BL, Cooke BaileyJN, Gaasterland D, Gaasterland T, Lee RK, Lichter PR,Budenz DL, Liu Y, et al. 2014. Association of CAV1/CAV2genomic variants with primary open-angle glaucomaoverall and by gender and pattern of visual field loss.Ophthalmology 121: 508–516.

Lu Y, Vitart V, Burdon KP, Khor CC, Bykhovskaya Y, Mir-shahi A, Hewitt AW, Koehn D, Hysi PG, Ramdas WD, etal. 2013. Genome-wide association analyses identify mul-tiple loci associated with central corneal thickness andkeratoconus. Nat Genet 45: 155–163.

Macgregor S, Hewitt AW, Hysi PG, Ruddle JB, Medland SE,Henders AK, Gordon SD, Andrew T, McEvoy B, Sanfi-lippo PG, et al. 2010. Genome-wide association identifiesATOH7 as a major gene determining human optic discsize. Hum Mol Genet 19: 2716–2724.

Manolio TA, Brooks LD, Collins FS. 2008. A HapMap har-vest of insights into the genetics of common disease. JClin Invest 118: 1590–1605.

Mears AJ, Jordan T, Mirzayans F, Dubois S, Kume T, ParleeM, Ritch R, Koop B, Kuo WL, Collins C, et al. 1998.Mutations of the Forkhead/Winged-Helix gene,FKHL7, in patients with Axenfeld-Rieger anomaly. AmJ Hum Genet 63: 1316–1328.

Michels-Rautenstrauss KG, Mardin CY, Zenker M, JordanN, Gusek-Schneider GC, Rautenstrauss BW. 2001. Pri-mary congenital glaucoma: Three case reports on novelmutations and combinations of mutations in the GLC3A(CYP1B1) gene. J Glaucoma 10: 354–357.

Mimiwati Z, Mackey DA, Craig JE, Mackinnon JR, Rait JL,Liebelt JE, Ayala-Lugo R, Vollrath D, Richards JE. 2006.Nail-patella syndrome and its association with glaucoma:A review of eight families. Br J Ophthalmol 90: 1505–1509.

Minegishi Y, Iejima D, Kobayashi H, Chi ZL, Kawase K,Yamamoto T, Seki T, Yuasa S, Fukuda K, Iwata T. 2013.Enhanced optineurin E50K–TBK1 interaction evokesprotein insolubility and initiates familial primary open-angle glaucoma. Hum Mol Genet 22: 3559–3567.

Morton S, Hesson L, Peggie M, Cohen P. 2008. Enhancedbinding of TBK1 by an optineurin mutant that causes afamilial form of primary open angle glaucoma. FEBS Lett582: 997–1002.

Munaut C, Lambert V, Noel A, Frankenne F, Deprez M,Foidart JM, Rakic JM. 2001. Presence of oestrogen recep-tor typeb in human retina. Br J Ophthalmol 85: 877–882.

Narooie-Nejad M, Paylakhi SH, Shojaee S, Fazlali Z, KanaviMR, Nilforushan N, Yazdani S, Babrzadeh F, Suri F, Ro-naghi M, et al. 2009. Loss of function mutations in thegene encoding latent transforming growth factor b bind-ing protein 2, LTBP2, cause primary congenital glauco-ma. Hum Mol Genet 18: 3969–3977.

Naumann GO, Schlotzer-Schrehardt U, Kuchle M. 1998.Pseudoexfoliation syndrome for the comprehensive oph-

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 11

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 12: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

thalmologist: Intraocular and systemic manifestations.Ophthalmology 105: 951–968.

Pang CP, Fan BJ, Canlas O, Wang DY, Dubois S, Tam PO,Lam DS, Raymond V, Ritch R. 2006. A genome-wide scanmaps a novel juvenile-onset primary open angle glauco-ma locus to chromosome 5q. Mol Vis 12: 85–92.

Pasquale LR, Kang JH. 2009. Lifestyle, nutrition and glau-coma. J Glaucoma 18: 423.

Pasquale LR, Loomis SJ, Weinreb RN, Kang JH, Yaspan BL,Bailey JC, Gaasterland D, Gaasterland T, Lee RK, ScottWK, et al. 2013. Estrogen pathway polymorphisms inrelation to primary open angle glaucoma: An analysisaccounting for gender from the United States. Mol Vis19: 1471–1481.

Ramdas WD, van Koolwijk LM, Ikram MK, Jansonius NM,de Jong PT, Bergen AA, Isaacs A, Amin N, Aulchenko YS,Wolfs RC, et al.. 2010. A genome-wide association studyof optic disc parameters. PLoS Genet 6: e1000978.

Ramdas WD, van Koolwijk LM, Ikram MK, Jansonius NM,de Jong PT, Bergen AA, Isaacs A, Amin N, Aulchenko YS,Wolfs RC, et al. 2011. Common genetic variants associ-ated with open-angle glaucoma. Hum Mol Genet 20:2464–2471.

Reis LM, Tyler RC, Volkmann Kloss BA, Schilter KF, LevinAV, Lowry RB, Zwijnenburg PJ, Stroh E, Broeckel U, Mur-ray JC, et al. 2012. PITX2 and FOXC1 spectrum of mu-tations in ocular syndromes. Eur J Hum Genet 20: 1224–1233.

Rezaie T, Child A, Hitchings R, Brice G, Miller L, Coca-Prados M, Heon E, Krupin T, Ritch R, Kreutzer D, et al.2002. Adult-onset primary open-angle glaucoma causedby mutations in optineurin. Science 295: 1077–1079.

Richards AJ, Yates JR, Williams R, Payne SJ, Pope FM, ScottJD, Snead MP. 1996. A family with Stickler syndrome type2 has a mutation in the COL11A1 gene resulting in thesubstitution of glycine 97 by valine in a1(XI) collagen.Hum Mol Genet 5: 1339–1343.

Riise R, Storhaug K, Brøndum-Nielsen K. 2001. Rieger syn-drome is associated with PAX6 deletion. Acta OphthalmolScand 79: 201–203.

Roh M, Zhang Y, Murakami Y, Thanos A, Lee SC, VavvasDG, Benowitz LI, Miller JW. 2012. Etanercept, a widelyused inhibitor of tumor necrosis factor-a (TNF-a), pre-vents retinal ganglion cell loss in a rat model of glaucoma.PLoS ONE 7: e40065.

Russo R, Cavaliere F, Watanabe C, Nucci C, Bagetta G, Cor-asaniti MT, Sakurada S, Morrone LA. 2008. 17b-Estradiolprevents retinal ganglion cell loss induced by acute rise ofintraocular pressure in rat. Prog Brain Res 173: 583–590.

Sarfarazi M, Stoilov I. 2000. Molecular genetics of primarycongenital glaucoma. Eye 14: 422–428.

Sarfarazi M, Akarsu NA, Hossain A, Turacli EM, Aktan GS,Barsoum-Homsy M, Chevrette L, Sayli SB. 1995. Assign-ment of a locus (GLC3A) for primary congenital glauco-ma (Buphthalmos) to 2p21 and evidence for genetic het-erogeneity. Genomics 30: 2171–2177.

Schlotzer-Schrehardt U. 2009. Molecular pathology of pseu-doexfoliation syndrome/glaucoma—New insights fromLOXL1 gene associations. Exp Eye Res 88: 776–785.

Schlotzer-Schrehardt U, Naumann GO. 1995. Trabecularmeshwork in pseudoexfoliation syndrome with and

without open-angle glaucoma. A morphometric, ultra-structural study. Invest Ophthalmol Vis Sci 36: 1750–1764.

Sethi A, Mao W, Wordinger RJ, Clark AF. 2011. Transform-ing growth factor–b induces extracellular matrix proteincross-linking lysyl oxidase (LOX) genes in human trabec-ular meshwork cells. Invest Ophthalmol Vis Sci 52: 5240–5250.

Sharafieh R, Child AH, Khaw PT, Fleck B, Sarfarazi M. 2013.LTBP2 gene analysis in the GLC3C-linked family and 94CYP1B1-negative cases with primary congenital glauco-ma. Ophthalmic Genet 34: 14–20.

Sheffield VC, Stone EM, Alward WL, Drack AV, Johnson AT,Streb LM, Nichols BE. 1993. Genetic linkage of familialopen angle glaucoma to chromosome 1q21-q31. Nat Ge-net 4: 47–50.

Sowers MR, Wilson AL, Kardia SR, Chu J, McConnell DS.2006. CYP1A1 and CYP1B1 polymorphisms and theirassociation with estradiol and estrogen metabolites inwomen who are premenopausal and perimenopausal.Am J Med 119: S44–S51.

Stein JD, Pasquale LR, Talwar N, Kim DS, Reed DM, Nan B,Kang JH, Wiggs JL, Richards JE. 2011. Geographic andclimatic factors associated with exfoliation syndrome.Arch Ophthalmol 129: 1053–1060.

Stoilov I, Akarsu AN, Sarfarazi M. 1997. Identification ofthree different truncating mutations in cytochromeP4501B1 (CYP1B1) as the principal cause of primarycongenital glaucoma (Buphthalmos) in families linkedto the GLC3A locus on chromosome 2p21. Hum MolGenet 6: 641–647.

Stoilov IR, Sarfarazi M. 2002. The third genetic locus(GLC3C) for primary congenital glaucoma (PCG)maps to chromosome 14q24.3. Invest Ophthalmol VisSci 43: E3015.

Stone EM, Fingert JH, Alward WL, Nguyen TD, PolanskyJR, Sunden SL, Nishimura D, Clark AF, Nystuen A, Nich-ols BE, et al. 1997. Identification of a gene that causesprimary open angle glaucoma. Science 275: 668–670.

Sud N, Wiseman DA, Black SM. 2010. Caveolin 1 is requiredfor the activation of endothelial nitric oxide synthase inresponse to 17b-estradiol. Mol Endocrinol 24: 1637–1649.

Swiderski RE, Reiter RS, Nishimura DY, Alward WL, Kale-nak JW, Searby CS, Stone EM, Sheffield VC, Lin JJ. 1999.Expression of the Mf1 gene in developing mouse hearts:Implication in the development of human congenitalheart defects. Dev Dyn 216: 16–27.

Thorleifsson G, Magnusson KP, Sulem P, Walters GB, Gudb-jartsson DF, Stefansson H, Jonsson T, Jonasdottir A, Jon-asdottir A, Stefansdottir G, et al. 2007. Common se-quence variants in the LOXL1 gene confer susceptibilityto exfoliation glaucoma. Science 317: 1397–1400.

Thorleifsson G, Walters GB, Hewitt AW, Masson G, Helga-son A, DeWan A, Sigurdsson A, Jonasdottir A, Gudjons-son SA, Magnusson KP, et al. 2010. Common variantsnear CAV1 and CAV2 are associated with primary open-angle glaucoma. Nat Genet 42: 906–909.

Toh T, Liew SH, MacKinnon JR, Hewitt AW, Poulsen JL,Spector TD, Gilbert CE, Craig JE, Hammond CJ, MackeyDA. 2005. Central corneal thickness is highly heritable:

R. Wang and J.L. Wiggs

12 Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 13: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

The twin eye studies. Invest Ophthalmol Vis Sci 46: 3718–3722.

Tokizane T, Shiina H, Igawa M, Enokida H, Urakami S,Kawakami T, Ogishima T, Okino ST, Li LC, Tanaka Y, etal. 2005. Cytochrome P450 1B1 is overexpressed and reg-ulated by hypomethylation in prostate cancer. Clin Can-cer Res 11: 5793–5801.

Tsuchiya Y, Nakajima M, Yokoi T. 2005. Cytochrome P450-mediated metabolism of estrogens and its regulation inhuman. Cancer Lett 227: 115–124.

Ueda J, Wentz-Hunter K, Yue BY. 2002. Distribution of my-ocilin and extracellular matrix components in the juxta-canalicular tissue of human eyes. Invest Ophthalmol VisSci 43: 1068–1076.

van Koolwijk LM, Ramdas WD, Ikram MK, Jansonius NM,Pasutto F, Hysi PG, Macgregor S, Janssen SF, Hewitt AW,Viswanathan AC, et al. 2012. Common genetic determi-nants of intraocular pressure and primary open-angleglaucoma. PLoS Genet 8: e1002611.

Vithana EN, Aung T, Khor CC, Cornes BK, Tay WT, Sim X,Lavanya R, Wu R, Zheng Y, Hibberd ML, et al. 2011.Collagen-related genes influence the glaucoma risk fac-tor, central corneal thickness. Hum Mol Genet 20: 649–658.

Vithana EN, Khor CC, Qiao C, Nongpiur ME, George R,Chen LJ, Do T, Abu-Amero K, Huang CK, Low S, et al.2012. Genome-wide association analyses identify threenew susceptibility loci for primary angle closure glauco-ma. Nat Genet 44: 1142–1146.

Volkmann BA, Zinkevich NS, Mustonen A, Schilter KF, Bo-senko DV, Reis LM, Broeckel U, Link BA, Semina EV.2011. Potential novel mechanism for Axenfeld-Riegersyndrome: Deletion of a distant region containing regu-latory elements of PITX2. Invest Ophthalmol Vis Sci 52:1450–1459.

Wang DY, Fan BJ, Canlas O, Tam PO, Ritch R, Lam DS, FanDS, Pang CP. 2004. Absence of myocilin and optineurinmutations in a large Philippine family with juvenile onsetprimary open angle glaucoma. Mol Vis 10: 851–856.

Wang DY, Fan BJ, Chua JK, Tam PO, Leung CK, Lam DS,Pang CP. 2006. A genome-wide scan maps a novel juve-nile-onset primary open-angle glaucoma locus to 15q.Invest Ophthalmol Vis Sci 47: 5315–5321.

Wiggs JL, Vollrath D. 2001. Molecular and clinical evalua-tion of a patient hemizygous for TIGR/MYOC. ArchOphthalmol 119: 1674–1678.

Wiggs JL, Del Bono EA, Schuman JS, Hutchinson BT, Wal-ton DS. 1995. Clinical features of five pedigrees geneti-cally linked to the juvenile glaucoma locus on chromo-some 1q21-q31. Ophthalmology 102: 1782–1789.

Wiggs JL, Allingham RR, Vollrath D, Jones KH, De La Paz M,Kern J, Patterson K, Babb VL, Del Bono EA, Broomer BW,et al. 1998. Prevalence of mutations in TIGR/Myocilin inpatients with adult and juvenile primary open-angleglaucoma. Am J Hum Genet 63: 1549–1552.

Wiggs JL, Lynch S, Ynagi G, Maselli M, Auguste J, Del BonoEA, Olson LM, Haines JL. 2004. A genomewide scanidentifies novel early-onset primary open-angle glauco-ma loci on 9q22 and 20p12. Am J Hum Genet 74: 1314–1320.

Wiggs JL, Kang JH, Yaspan BL, Mirel DB, Laurie C, Cren-shaw A, Brodeur W, Gogarten S, Olson LM, Abdrabou W,et al. 2011. Common variants near CAV1 and CAV2 areassociated with primary open-angle glaucoma in Cauca-sians from the USA. Hum Mol Genet 20: 4707–4713.

Wiggs JL, Kang JH, Yaspan BL, Mirel DB, Laurie C, Cren-shaw A, Brodeur W, Gogarten S, Olson LM, Abdrabou W,et al. 2012. Common variants at 9p21 and 8q22 are asso-ciated with increased susceptibility to optic nerve degen-eration in glaucoma. PLoS Genet 8: e1002654.

Wiggs JL, Hauser MA, Abdrabou W, Allingham RR, BudenzDL, Delbono E, Friedman DS, Kang JH, Gaasterland D,Gaasterland T, et al. 2013a. The NEIGHBOR Consortiumprimary open-angle glaucoma genome-wide associationstudy: Rationale, study design, and clinical variables. JGlaucoma 22: 517–525.

Wiggs JL, Howell GR, Linkroum K, Abdrabou W, Hodges E,Braine CE, Pasquale LR, Hannon GJ, Haines JL, John SW.2013b. Variations in COL15A1 and COL18A1 influenceage of onset of primary open angle glaucoma. Clin Genet84: 167–174.

Winnier GE, Kume T, Deng K, Rogers R, Bundy J, Raines C,Walter MA, Hogan BL, Conway SJ. 1999. Roles for thewinged helix transcription factors MF1 and MFH1 incardiovascular development revealed by nonallelic non-complementation of null alleles. Dev Biol 213: 418–431.

Yam GH, Gaplovska-Kysela K, Zuber C, Roth J. 2007. So-dium 4-phenylbutyrate acts as a chemical chaperone onmisfolded myocilin to rescue cells from endoplasmic re-ticulum stress and apoptosis. Invest Ophthalmol Vis Sci48: 1683–1690.

Ying H, Yue BY. 2012. Cellular and molecular biology ofoptineurin. Int Rev Cell Mol Biol 294: 223–258.

Zhou X, Li F, Ge J, Sarkisian SR, Tomita H, Zaharia A,Chodosh J, Cao W. 2007. Retinal ganglion cell protectionby 17-b-estradiol in a mouse model of inherited glauco-ma. Dev Neurobiol 67: 603–616.

Zhou YY, Zhao Y, Mirza MK, Huang JH, Potula HHSK,Vogel SM, Brovkovych V, Yuan JXJ, Wharton J, MalikAB. 2009. Persistent eNOS activation secondary to cav-eolin-1 deficiency induces pulmonary hypertension inmice and humans through PKG nitration. J Clin Invest119: 2009–2018.

Zode GS, Clark AF, Wordinger RJ. 2009. Bone morphoge-netic protein 4 inhibits TGF-b2 stimulation of extracel-lular matrix proteins in optic nerve head cells: Role ofgremlin in ECM modulation. Glia 57: 755–766.

Zode GS, Kuehn MH, Nishimura DY, Searby CC, Mohan K,Grozdanic SD, Bugge K, Anderson MG, Clark AF, StoneEM, et al. 2011. Reduction of ER stress via a chemicalchaperone prevents disease phenotypes in a mouse mod-el of primary open angle glaucoma. J Clin Invest 121:3542–3553.

Zode GS, Bugge KE, Mohan K, Grozdanic SD, Peters JC,Koehn DR, Anderson MG, Kardon RH, Stone EM, Shef-field VC. 2012. Topical ocular sodium 4-phenylbutyraterescues glaucoma in a myocilin mouse model of primaryopen-angle glaucoma. Invest Ophthalmol Vis Sci 53:1557–1565.

Common and Rare Genetic Risk Factors for Glaucoma

Cite this article as Cold Spring Harb Perspect Med 2014;4:a017244 13

ww

w.p

ersp

ecti

vesi

nm

edic

ine.

org

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from

Page 14: Common and Rare Genetic Risk Factors for Glaucomaperspectivesinmedicine.cshlp.org/content/4/12/a017244.full.pdf · genes are responsible for congenital glaucoma (CYP1B1andLTBP2),developmentalglaucoma

September 18, 20142014; doi: 10.1101/cshperspect.a017244 originally published onlineCold Spring Harb Perspect Med 

 Ryan Wang and Janey L. Wiggs Common and Rare Genetic Risk Factors for Glaucoma

Subject Collection Retinal Disorders: Genetic Approaches to Diagnosis and Treatment

Degenerations in Systemic DiseaseA Review of Secondary Photoreceptor

Naveen Mysore, Jamie Koenekoop, Shen Li, et al.

What Is Next for Retinal Gene Therapy?Luk H. Vandenberghe

Dominant Retinitis PigmentosaGenes and Mutations Causing Autosomal

SullivanStephen P. Daiger, Sara J. Bowne and Lori S.

and Efficacy Gene Therapy Trials: SafetyRPE65The Status of

Eric A. Pierce and Jean Bennett

Retinal Biology and DiseaseRNA-Seq: Improving Our Understanding of

Sousa, et al.Michael H. Farkas, Elizabeth D. Au, Maria E.

RetinoschisisStudies: Gene Therapy for X-Linked Convergence of Human Genetics and Animal

Ronald A. Bush, Lisa L. Wei and Paul A. Sieving

Macular DegenerationDifferential Gene Expression in Age-Related

Denise J. Morgan and Margaret M. DeAngelisMacular DegenerationGene Therapies for Neovascular Age-Related

ScariaPeter Pechan, Samuel Wadsworth and Abraham

Optic Nerve: Clinical ApplicationsGenetics of Primary Inherited Disorders of the

Keri F. Allen, Eric D. Gaier and Janey L. WiggsMYO7AUsher Syndrome Caused by Mutations in

Gene Therapy for the Retinal Degeneration of

Vanda S. Lopes and David S. WilliamsGenetic Modifiers and Oligogenic Inheritance

Maria Kousi and Nicholas Katsanis-Associated DiseasesABCA4Gene Therapy of

AllikmetsAlberto Auricchio, Ivana Trapani and Rando

Inherited Retinal DegenerationsStem Cells as Tools for Studying the Genetics of

et al.Luke A. Wiley, Erin R. Burnight, Robert F. Mullins,

Gene Therapy Using Stem Cells

et al.Erin R. Burnight, Luke A. Wiley, Robert F. Mullins,

RPGRIP1in Leber Congenital Amaurosis Caused by Mutations

Tiansen LiAdeno-Associated Viral (AAV) VectorGene Therapy for Choroideremia Using an

MacLarenAlun R. Barnard, Markus Groppe and Robert E.

http://perspectivesinmedicine.cshlp.org/cgi/collection/ For additional articles in this collection, see

Copyright © 2014 Cold Spring Harbor Laboratory Press; all rights reserved

on July 4, 2020 - Published by Cold Spring Harbor Laboratory Press http://perspectivesinmedicine.cshlp.org/Downloaded from


Recommended