Open access peer-reviewed chapter

Molecular Genomics of Glaucoma: An Update

Written By

Govindasamy Kumaramanickavel, Siddhita Jadhav, Sugirdhana Parthiban Ramsait and Pinakin Gunvant Davey

Submitted: 09 August 2022 Reviewed: 21 October 2022 Published: 09 December 2022

DOI: 10.5772/intechopen.108697

From the Edited Volume

Glaucoma - Recent Advances and New Perspectives

Edited by Pinakin Gunvant Davey

Chapter metrics overview

198 Chapter Downloads

View Full Metrics

Abstract

Glaucoma is in the top five age-related eye disorders with increasing prevalence globally. Past research has led to the understanding of glaucoma as a neurodegenerative disease. Glaucoma phenomics could be syndromic or non-syndromic. Globally primary open angle, primary angle closure and primary pseudoexfoliation glaucomas are widely present. The genetics and genomics of glaucoma are heterogeneous, both clinically and genetically. Glaucoma has heritability associations, particularly with central corneal thickness, retinal nerve fibre layer and peripapillary atrophy. Ocular embryogenesis genes when mutated could cause either local (in situ), pan-ocular or systemic syndromic glaucoma phenomics. In glaucoma, except for a few single gene causes, most of the associations have been shown with innumerable gene single-nucleotide polymorphisms and epigenetic factors. The biological mechanisms in glaucoma are mechanical strain, inflammation, oxidative stress, vascular dysregulation, and immune imbalance, which independently or collectively contribute to the neurodegeneration and visual morbidity. Biomarkers in glaucoma have experimental study biases and therefore today we cannot apply them effectively in clinical practice and henceforth that demands further research to understand the fundamental basis of the disease. However, the knowledge gained in research will translate into early detection and biomolecular interventional strategies, having traction toward personalised medicine.

Keywords

  • age-related eye diseases
  • biomarkers
  • biomechanisms
  • epigenetics
  • glaucoma
  • genomewide associations
  • genetics
  • gene therapy
  • neurodegeneration
  • neuroprotection
  • personalised medicine

1. Introduction

Genetics is usually the study of a gene and the corresponding physiological trait or disease phenotype, which is inherited through generations; whereas genomics is the study of all the genes, genes expressed in the person’s genome which are responsible for a physiological or pathophysiological phenotype (phenomics) in the health or disease. The phenotype of glaucoma is heterogeneous, varying from a spectrum of normal tension, high tension to retinal ganglion cell death and visual morbidity. Similarly, the molecular genomics of glaucoma is complex, which is unlike corneal, lens or retinal genomics, where the seat of the disease is localised to the site of the respective tissues. However, in contrast glaucoma is pan ocular – extending from the anterior segment to posterior segment of the eye and the optic nerve, and thus, several anatomical regional tissues of the eye and genes, gene expressions are the stakeholders in the molecular mechanism of glaucoma. In addition, the disease outcome is measurable in the tears, aqueous humour, ciliary body, trabecular meshwork, vitreous body, lamina cribrosa (superficial nerve fibre layer, retinal ganglion cells, prelaminar region, laminar region, retrolaminar region), retina, optic nerve, serum and blood, which collectively blurs a single cause and effect of the glaucoma machinery [1, 2]. At the same time, these candidates remain to be the barriers and opportunities in glaucoma screening measures, early clinical detection, effective clinical management, valuable prognostication and futuristic molecular interventions.

The genetics of glaucoma is less of Mendelian and more of complex nature, perhaps more diverse compared to any other age-related eye diseases (ARED), like for example, age-related macular degeneration (AMD). In glaucoma genomics, there are very few genes which behave as a Mendelian single gene disease, while several genes and single nucleotide polymorphisms (SNPs), gene expression modulations, correspond to the pathophysiological traits as a neurodegenerative disease. There are a couple of hundred genes, several hundred SNPs, and many microRNAs which all are associated with ARED glaucoma phenomics. Many of the genomewide association studies are robust, where large collaborative sample sizes, validation studies, across different populations have been designed, executed and published. The molecular mechanisms of glaucoma are a spectrum of clinical outcomes played by several biological actors, beginning from inflammation, oxidative stress, extracellular matrix dysregulation, immune system imbalance, neuroprotection, neurodegeneration, apoptosis, metabolites accumulation, to abnormal lipid factors. However, most of the molecular genomic factor studies are not robust and are unfortunately poorly validated. Besides, glaucoma manifests in the elderly as a result of mix and match with other AREDs visual morbidities like cataract, corrected or uncorrected refractive errors, AMD, and diabetic retinopathy and therefore usually may not be isolated. For example, a person with glaucoma may have cataracts and/or AMD as well, again this could be another bias factor in the molecular genomics laboratory studies. Nevertheless, in this review, we shall have an in-depth overview of the molecular genetics and genomic factors associated with the pathophysiological mechanisms of glaucoma. However, the review also provides a larger insight into the visual impairment, prevalence, and comorbidities, besides the genetics and genomics of glaucoma. However, it is beyond the scope of the review to provide a gist of all the biological, experimental, epidemiological, genetic and genomic studies in glaucoma and hence, kindly refer to the references provided at the end, for further information.

Advertisement

2. Glaucoma in general

2.1 Visual impairment, age-related disorders and the central role of glaucoma

In the elderly, glaucoma cannot be viewed as an isolated pathology and it is frequently associated with other age-related visual and systemic comorbidities like ocular (cataracts, age-related macular degeneration, type 2 diabetes mellitus and its complications, visual impairments, diabetic retinopathy) and non-ocular (airways diseases, coronary artery disease, hypertension, heart failure, dementia, depression, et cetera). Besides, the treatment of glaucoma will have an effect on some of the morbidities mentioned [3]. As the population of the aged increase, common causes of visual morbidity increase significantly. In 2015, the three top causes of blindness were preventable - cataract, uncorrected refractive error (URE) and glaucoma, whereas for visual impairment it is URE, cataract and AMD. Effective and largescale eyecare service is required to combat these problems [4]. In 2017, Ackland et al., published that 253 million people are visually impaired with 217 million moderately or severely visually impaired (MSVI) and 36 million blind and they estimate that by 2050 these numbers would climb sharply to 588 million MSVI and 115 million blind globally [5]. About 89% of the VI live in low or middle-income countries and 55% of them are women. About 1.1 billion people have uncorrected functional presbyopia. Though the prevalence of VI has reduced from 4.58% to 3.38%, more thrust has to be given to reduce it further [5]. Eckert et al., estimated the cost of blindness as US$ 7.8 billion in the US and the cost of MSVI as US$ 16.5 billion, however, Gordois et al., estimated the cost of VI as a staggering US$ 3 trillion and the direct costs as US$ 2.3 trillion and also mentions that these figures could increase by 20% by the year 2020 [6]. Another non-profit organisation of Prevent Blindness in America estimates that the economic burden of adult vision problems (AMD, cataract, diabetic retinopathy, glaucoma, refractive errors, VI and blindness) in the US in 2007 as US$ 54.1 billion annually, which includes direct medical, direct-other and loss of productivity costs, however, they revised the figure to US$ 139 billion in a 2013 report [7].

In 2015, it was estimated that the moderate to severe VI affected 216.6 million globally, with the URE being the leading cause, and blindness prevalent amongst 36 million with cataract outnumbering the other causes [4]. The proportion of those with preventable or treatable blindness and VI is reducing in trend over the decades, fortunately, as mentioned earlier, due to the control of infectious and nutritional causes. And amongst those with visual morbidity, aged individuals comprise the maximum, having the distribution variable between the developed and developing countries [8, 9]. ARED, such as AMD, cataract, diabetic retinopathy (DR), glaucoma and refractive errors are the key components of global visual morbidity and cataract forms more than half of all those affected in the group. Amongst those, 70–74 years of age 37% have cataract, 10% AMD, 3% glaucoma and 2% DR [10]. Out of the 285 million with VI and blindness, those above 50 years of age constitute 65% of the VI and 82% of blindness. In addition, due to poor socio-economic status and biological element like longevity, 75% of those affected with ARED are women and this factor is consistent irrespective of the fact that whether women live in developed or developing countries [11, 12].

In Germany, ARED commonly found were cataract, dry eye, AMD and glaucoma, furthermore, they found that the aged individuals had different combinations of these conditions [13]. Asia has one of the highest representations of the blind, with India having the highest prevalence of 11.9% and Malaysia with the lowest of 0.3% [14]. Due to the robust epidemiological studies, ARED in Asia includes, along similar lines to that of the West, cataract, refractive errors, glaucoma, DR and AMD. AMD is prevalent in both the developed and developing countries as being the cause of blindness and VI, the prevalence of the disease is higher in the West, but the emerging trends and patterns from China, India, Japan, Mongolia, Singapore to Taiwan are echoing the West, due to the growing aged populations [14]. ARED in Iran had a similar pattern to most Asian countries with 35.8% having either cataract, AMD, glaucoma or DR and moreover one in two of those over 75 years of age have these conditions [15]. In the US, a third of the subjects were either 80 years or over who had cataract, AMD, primary open-angle glaucoma (POAG), DR or VI and two-thirds had late AMD. In addition, POAG, VI and DR were prevalent at a higher age amongst Hispanics and Blacks, whereas cataract and late AMD prevalence were higher amongst the Whites [16]. The prevalence of ARED in Canada increased alarmingly after the age of 75 years [17]. In high-income Eastern and Central European countries, blindness and mild to severe VI reduced between 1990 and 2015 from 0.26% to 0.15% and from 1.74% to 1.27%, respectively and similar trends were observed in Australasia, North America and Western Europe. One in 28, above 40 years have low vision in the USA [18]. Conflicting reports are available in the US demonstrating that vision screening methods could improve the visual status of a community, in older adults [19]. In Britain, Prasad et al., observed that diabetes was not the primary factor for the prevalence of blindness and MSVI, whereas non-diabetes factors were particularly responsible [20, 21]. In Latin America for the elderly, 50 years of age or above, the prevalence of blindness varied from 1.1% in Argentina to 4.4% in Guatemala, with cataract being the foremost reason, however, DR and glaucoma are rising and infectious diseases are declining [22]. In the population of Indian origin in Singapore, 40 years or above, the prevalence of VI and blindness were 3.4% and 0.4% respectively, far lower than in India, for which cataract, DR, AMD and glaucoma were the leading causes and the first was the primary cause [23, 24]. ARED in Singapore Indians 40 years or above irrespective of education level, literacy or immigration types were deteriorating and active screening measures should be implemented rather than voluntary enrolment is emphasised [25].

With the ageing population, rising in proportion across the world despite the fall in birth rates, the corresponding increase in morbidity and mortality amongst the group is worrying. Universal health coverage and eye health objectives are persevering to reduce global visual morbidity for which robust databases are key to achieving the goals [26, 27, 28]. Biological understanding of ARED, not only clinical screening, is equally important for the prevention and management of the diseases. Oxidative stress and inflammation are the key causative mechanisms for ARED. Autophagy mechanisms also play both protective and detrimental outcomes in ARED and therefore nurturing preventive and therapeutic strategies [29]. Malnutrition and anaemia have been associated with poor vision besides other systemic disorders in the elderly, in a study from southern India [30]. In a southern Indian glaucoma study, primarily around three-quarters were due to cataract and the remaining were because of glaucoma, cystoid macular oedema, optic atrophy and corneal scars and these were significantly associated with ageing (p < 0.0001) [31].

2.2 Prevalence of glaucoma

Globally, the five leading causes of visual impairment are, URE, cataract, AMD, glaucoma and DR. However, glaucoma is the second leading cause of loss of vision in the world. About 60.5 million people are estimated to be affected globally by glaucoma in 2010, which is equivalent to the population of Italy, and about 8.4 million of them will be suffering from bilateral irreversible blindness and there are closely varying estimates according to other studies [32]. These figures could rise to 111.8 million in 2040 and the global prevalence of glaucoma presently is around 3–4% [33, 34]. POAG is highest amongst African and Hispanic races and is found amongst all races, whereas primary angle closure glaucoma (PACG) is the highest in Asia [35, 36]. Primary congenital glaucoma (PCG) is a less common type, however primary exfoliative glaucoma (PXFG) is a comparatively more frequent disorder resulting in most common glaucoma related blindness, rather than the other types. In a focused metanalysis of five glaucoma prevalence studies in India by Geroge et al., it was estimated that in 2010 about 11.2 million would be affected with the disease and out of which 6.48 million would have POAG and 2.54 million, PACG and these figures should have increased in the decade that has passed since the publication [37]. The economic burden of glaucoma in the United States calculated by Rein et al., in 2006 was around US$ 2.9 billion, in this context we should take note that the majority of glaucoma cases are undiagnosed [38]. Studying five major prevalence studies in India, the age-standardised prevalence ranges of those 40 or 50 years or above with POAG was—1.29% to 4.24% and for PACG—0.5% to 1.11% and the reason for the wide range of variations are largely due to disparities in clinical and epidemiological study methodologies. Childhood glaucoma is constituted by primary congenital glaucoma and juvenile open angle glaucoma, which affects 1 in 10,000 to 100,000 children worldwide and the former is more prevalent in high consanguineous marriage geographical regions [33]. Pigmentary glaucoma or pigment dispersion syndrome is caused by PMEL gene variants only less than 50% of those with the variants get affected. PMEL is involved in melanin pigment synthesis, storage and transport and these pigments get deposited in the trabecular meshwork and increase the IOP.

Advertisement

3. Genetics and genomics of glaucoma

‘Glaykoseis’, a blindness in the elderly, as mentioned by Hippocrates—the Father of Modern Medicine, dates back to 400 years before Christ emerged and Amida, a Byzantine physician, named it as ‘Amaurosis’ [39]. Before 1850, POAG was termed as amaurosis, black cataract or gutta serena. The eyes were observed to be hard and angle-closure glaucoma caused green or grey pupils and hence the name glaucoma (blue, green or grey and viriditate occuli) and in 1850 after the ophthalmoscope invention, the scenario changed, when the term ‘Glaucoma’ was christened to the disease, which has not changed until today [39]. Ganglionic optic neuropathy is the pathological defect of glaucoma which leads to a painless visual loss. The molecular genetics and medical biology of glaucoma have intrigued scientists for a while, however, whatever knowledge that we have gained today is not yet as clear as compared to the inherited retinal degenerative diseases (IRDD).

3.1 Heritability of glaucoma

In the earlier days, twin studies were the proof to establish if a disease is caused by heritable or environmental factors. The twin studies in 1987 established that the heritability of POAG at 0.135 [40]. In addition, two recent robust studies with genomewide array data though parked the heritability of POAG between a wide range of 24–42% [41, 42]. The risk factors for developing glaucoma are age, ethnic origins (African Americans, Hispanics), gender (women), genetics, hypertension and increased intraocular pressure prescription drugs [43]. The prevalence of POAG was highest amongst the African race descent, then Asians, but the lowest was amongst the Europeans, showing racial and genetic preponderance. However, the genetics of the disease is complex with only 10% having Mendelian inheritance.

Central corneal thickness, intra-ocular pressure (IOP), optic disc area and vertical cup/disc ratio (VCDR) have high heritability associations across populations. Asefa et al., looked at the anterior chamber size, central corneal thickness (CCT), corneal hysteresis, cup-to-disc ratio, cup-shape, cup-size, disc-size, intraocular pressure, peripapillary atrophy (PA) and retinal never fibre layer thickness (RFNLT) in a metanalysis [44]. And the highest heritability was observed in CCT (h2 = 0.81), RFNLT (h2 = 0.73) and PA (h2 = 0.73) [44].

3.2 Mendelian genetics of glaucoma

There are very few single gene defect causations in non-syndromic glaucoma, so far, four PCG loci have been located—GLC3A, B, C and D. The GLC3A region in 2p21 has the CYP1B1 gene and about 150 or so autosomal recessive mutations have been associated with PCG. The majority of patients with PCG/CYP1B1 mutations are found in Saudi Arabia and Slovakia gypsy population and these mutations have variable expressivity and incomplete penetrance with a wide range of clinical phenotypes [45]. PCG is present across the world and being an autosomal recessive disease, is more frequent in consanguineous populations, has a widely varying incidence of 1 in 1250 to 1 in 22,000 in different parts of the world and is one of the paediatric causes of blindness in India [46, 47, 48]. A consanguineous south Indian family with PCG was investigated by using homozygosity analysis. The microsatellite markers D2S177 and D2S1346 were tightly linked to CYP1B1 and the Q110X mutation in exon 2 of the gene was co-segregating in all the affected [49]. A newborn in the family was found to be a heterozygous carrier to the relief of the family during genetic counselling [45]. CYP1B1 plays a critical role in the development of the trabecular meshwork (TM) and acts in the removal of reactive oxygen species, regulating oxidative stress and production of periostin which influences the mechanical strength and structural integrity of the TM [45]. GLC3B and C have not resulted in any gene till date, but GLC3D location at 14q24 has resulted in null mutations in LTBP2 and PCG phenotype correlation. LTBP2 (latent transforming growth factor beta binding protein 2) is a matrix protein that performs cell adhesion and tissue repair processes.

Optineurin (OPTN) gene mutation is one of the causes of POAG in various populations, though it is rate. In OPTN, M98K variation is associated with POAG across populations and in our study, was less frequent cause of the disease as it was found that in 4% of HTG and 6% of NTG patients compared to the controls [50]. OPTN plays critical role in Golgi complex maintenance, membrane trafficking, exocytosis, interacts with myosin VI and Rab8.

A sensational MYOC gene location for glaucoma was identified by Stone et al. in 1997 and is one of the most investigated in glaucoma genetics [51]. The sensational was though short-lived in the history of genetics of glaucoma and it lost its steam as the frequency of the mutations started to fall to very small proportions amongst the global glaucoma populations. When we screened 100 POAG/JOAG patients for the MYOC gene, we found a G144A, Gln48His substitution, which was novel at that time, in 2% of the patients, the change was also found in another four affected members of a JOAG family. MYOC was found to not play a significant role amongst glaucoma patients in India [52]. Juvenile open angle glaucoma (JOAG) was described in a large pedigree in the US seventy years ago and today the Phe369Leu MYOC mutation was identified in the family [53].

3.3 Molecular genetics of syndromic glaucoma

Syndromic glaucoma, which are also Mendelian in nature, may not be uncommon in the paediatric age group, hence glaucoma may be associated with additional ocular and systemic phenotypes like aniridia, anterior segmental dysgenesis, collagen or vascular disorders, immunogenetic diseases, metabolic disorders and nanophthalmos. The embryogenesis of the eye is complex with both the ectoderm and neuroectoderm involved together in the formation. Mutation in genes, infections acquired during or after pregnancy, ageing and systemic disorders affect the development of the eye [45]. Axenfeld-Rieger syndrome, a disorder is prevalent in 1:200,000 individuals, which affects the anterior segment like defective cornea, iris and the extraocular features include facial, dental and skeletal abnormalities. Peters’ anomaly, an autosomal dominant disorder, is another common condition affecting the anterior segment development where cornea, iris and lens resist to separate, leading to central corneal opacity and non-ocular features like cleft lip/palate, short stature, physical and mental retardation. Aniridia is another developmental condition accompanied by photophobia and poor visual acuity. The genetics of syndromic glaucoma is Mendelain.

WAGR syndrome with Wilms tumour, aniridia, genitourinary anomalies and intellectual disability earlier mentioned as mental retardation. In WAGR syndrome there will be chromosomal deletion at the 11p13 region which harbours many genes including mutated WT1 and/or PAX6 genes and the affected have glaucoma as well. Hence, aniridia patients should undergo an abdominal ultrasound to rule out WAGR syndrome and renal tumour. There is a rare condition of Gillespie syndrome, where patients have aniridia, ptosis and corectopia with mutation in the ITPR1 gene [54].

Collagen vascular diseases like Stickler syndrome and osteogenesis imperfecta patients have glaucoma due to trabecular meshwork impedance. Stickler syndrome patients have myopia, cataract and retinal giant tear, the iris ciliary process is long and covers the trabecular meshwork blocking the aqueous flow. The gene variants in COL2A1 and COL11A1 cause autosomal dominant Stickler syndrome, whereas variations in COL9A1 cause autosomal recessive type. Osteogenesis imperfecta has COL1A1 and COL1A2 gene variants manifesting in an autosomal dominant manner. Osteogenesis imperfecta is a collagen bone disorder with a variable five phenotypes, all mostly have low mineral density leading to bone fragility (hence the name brittle-bone disease), blue sclera, abnormal cornea, glaucoma, poorly formed dentine, the ligaments are hyper lax, cardiovascular disease and hearing loss [55]. Immune-related disorders like Aicardi-Goutieres syndrome (AGS) and Singleton-Merten syndrome (SGMRT), are severe and fatal conditions with a plethora of genes involved in RNA processing like ADAR, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, TREX1 and the gene IFIH1 responsible for innate immunity. In a severe type, patients could have cerebral atrophy, congenital glaucoma, hepatosplenomegaly, intracranial calcification, leukodystrophy, microcephaly, thrombocytopenia and death are not uncommon. Singleto-Merten syndrome is caused by variants in the DDX58 and IFIH1 genes and has glaucoma as a clinical feature.

Nanophthalmos, with small fully formed eyes, could be inherited as both autosomal dominant and recessive types. There are several genes involved in this condition (CRB1, BEST1, FAM111A, MFRP, MYRF, PRSS56 and TIMEM98), which has additional systemic features like congenital diaphragmatic hernia, cardio-pulmonary abnormalities, glaucoma and urogenital anomalies, causing some rare genetic disorders like Kenny-Caffey syndrome [56]. MYRP, CRB1 and BEST1 are genes associated with retinal degenerative genetic disorders. TEK/ANGPT1 genes’ variants independently resulted in haploinsufficiency-based primary congenital glaucoma. The genes have a critical role in the structure and function of Schlemm’s canal and trabecular meshwork [57].

PITX2 (Paired Like Homeodomain 2) in chromosome 4q25 and FOXC1 Forkhead Box C1 are transcription factors jointly involved in the anterior segment development. Mutations or copy number variations in these genes result in anterior segment anomalies, due to haploinsufficiency, like Axenfeld Reiger syndrome or Peters’ anomaly [58]. Axenfeld first described the anomaly in 1920 and later was added more by Reiger in 1934 [56]. A majority of the children with mutations in PITX2 and FOXC1 develop glaucoma [56]. Non-ocular systemic features include variable phenotypes like facial/dental anomalies, pituitary involvement, umbilical anomalies, syndromes like SHORT, short FRAME, cardiac defects, sensorineural deafness and myotonic dystrophy [56]. The development of the eye is highly complicated and well-studied in Drosophila and humans [59]. PAX6 gene in humans or ey gene in drosophila is the chief conductor of the biological symphony of eye development. PAX6 (Transcription factor Paired Box 6) gene in chromosome 11p13 mutation is associated with mostly aniridia but rarely Peters’ anomaly and other ocular defects have also been reported [58]. Peters’ anomaly could be caused by a variety of genes like PAX6, PITX2, FOXC1, CYP1B1 and B3GALTL as they all collectively during embryogenesis orchestrate the anterior segment development [56].

FOXC1 (forkhead box) gene mutation and haploinsufficiency cause anterior segment anomalies [59]. The MAF basic region leucine zipper (bZIP) transcription factors perform anterior segment and lens development and mutations in the gene in humans cause cataract and ocular developmental defects. CPAMD8 (C3 and PZP-like alpha-2-macroglobulin domain-containing protein 8) mutation causes anterior segment dysgenesis (ectropion uveae, cataract, corectopia, iridodonesis with ectopia lentis) in an autosomal recessive manner [56]. CPAMD8 is involved in the dynamics of aqueous humour.

B3GALTL (Beta-3-Glucosyltransferase) gene in 13q12.3 which glycosylates proteins and when mutated causes Peter Plus syndrome manifesting with ocular and systemic features like abnormal ears, brachydactyly, cleft lip/palate, dextrocardia, dysmorphic face, hydrocephalus and Potter syndrome [56]. SOX2 (SRY-like box2) involved in eye development is mapped to 3q26.3-27 and mutations in the gene cause sclerocornea and anophthalmia [60]. CHRDL1 (Chordin like 1) mutation causes megalocornea and Neuhauser syndrome and the gene is X-linked and located in the Xq23 region [61]. Keratoconus, myopia and glaucoma are associated with glaucoma and hence mutations in the autosomal dominant gene associated with the former is the VSX1 (Visual System Homeobox 1), a transcription factor located in 20p11.21 [62]. Genes like COL4A1, CYP1B1, FGFR2, BMP4, BMP7, FOXE3, MYOC LAMBB2 and LTBP2 are involved in anterior segment anomalies [63].

SIX3 gene mutation causes holoprosencephaly or microphthalmia and iris coloboma [59]. SIX3 interacts with Groucho-related proteins 4 and 5 and functions as an eye development repressor. Besides, PAX6 and SIX3 regulate each other during eye development. Along with FOXE3, MAF, MITF, LHX2, PITX3, PROX1, and SIX3, PAX6 forms the cornea and lens, whereas along with CHX10, EYA1 and PAX2 forms the retina and optic nerve. In addition, genes like BMP4, BMP7, RX and SHH also regulate PAX6 in eye development and mutations in them affects eye development which may result in glaucoma.

In metabolic disorders, the X-linked recessive and autosomal recessive mucopolysaccharidoses [Hurler syndrome (alpha-L-iduronidase), Hunter syndrome (iduronate2-sulfatase), Sanfilippo syndrome (heparin sulphate), Morquino syndrome (N-acetyl galactosamine-6-sulfatase), Maroteauz-Lamy syndrome (N-acetyl galactosamine-4-sulfatase), Natowicz syndrome (hyaluronidase)] gene mutations could result in defective enzyme causing cataract and glaucoma and the latter by blocking the trabecular meshwork with the glycosaminoglycans [56].

Advertisement

4. Complex genetics and genomics of glaucoma

Syndromic glaucoma is well understood genetically and genomically, with the fundamental knowledge of the cause and effect. However, POAG is a multifactorial disease, where embryological development, genetics, epigenetics, genetic polymorphisms, variable gene expressivity, inflammation and environmental modifiers play a collective complex role and in addition, the penetrance and expressivity may vary between affected individuals [64]. All mentioned earlier, is applicable to most of the lifestyle related complex genetic disorders. This means that the person with a genetic risk may or may not manifest the disease and hence, it is completely unlike the Mendelian genetics. Hence, the disease aetiology, onset, duration, drug response and inheritance are dependent on genetics and environmental modifiers. In addition, some non-genetic modifiers complicate the glaucoma disease status, like smoking, and comorbidities (diabetes, untreated high blood pressure) and near-sightedness [64].

Genetic Epidemiology Research in Adult Health and Ageing (GERA) is part of the UK Biobank (UKB) that has phenotype and genotypes of 500,000 participants aged 40–69 years, which has multi-ethnic glaucoma cases of 7329 and 169,561 controls [65]. Choquet et al., in a GERA study, having 4986 POAG cases and 58, 426 controls comprising of African-Americans, non-Hispanic whites, Hispanic/Latinos, and East-Asian races and ethnicities, identified 24 loci for POAG, out of which 14 were novel and 9 replicated near the genes FMNL2, PDE7B, TMTC2, IKZF2, CADM2, DGKG, ANKH, EXOC2, and LMX1B, across races, but was found higher in African-Americans. Some of the genes had functional influence like FMNL2 and LMX1B – Lmx1b mutations increase the IOP and POAG in mice. A metanalysis of GERA and UKB further identified 24 additional loci expanding the spectrum of the genetics of POAG, however, most of the variants have minimal genetic risk [66]. Burdon et al., have associated the following genes with ocular physiological traits, which are key in maintaining the IOP in POAG - ZNF469, FOXO1, COL5A1, AKAP13, AVGR8, COL8A2, IBTK, LRRK1/CHSY1, C7orf42, ATOH7, TGFBR3, CARD10, CDC7/TGFBR3, SALL1, CDKN2A/B, SIX1/SIX6, FERM8/SCYL1, DCLK1 and CHEK2 [67]. Furthermore, POAG associated candidate genes have been identified, CAV1/CAV2, TMCO1, CDKN2B-AS1, TXNRD2, ATXN2, FOXC1 and GAS7 [68, 69]. Some of the genes are consistent across various studies besides ATOH7, CAV1/CAV2, CDC7-TGFBR3, CDKN2B-AS1, GAS7, SIX1/SIX6 and TMCO1; these are not only associated with POAG but also with the quantitative traits (endophenotypes) [68]. However, some genes having mutations do affect a small proportion of those with POAG, such as cyclin-dependent kinase inhibitor 2B, myocilin (MYOC), neurotrophin 4, optineurin (OPTN), tank binding kinase 1 (TBK1) and WDR 36. Other types of glaucoma like PXFG are associated with LOXL1 and CNTNAP2 and PCG with CYP1B1 and LTBP2 [69, 70]. MYOC, OPTN and TBK1 are used in genetic diagnosis, counselling and clinical management, in addition to this list even CYP1B1 could be added [69]. Verma et al., in a complex gene–gene interaction modelling using NEIGHBOUR, eMERGE datasets and tissue expressing databases identified a new set of genes like GNG7, ROBO1, SUMF1, RYR3, SLC24A3, CCDC3, CARS2, RPS6KA, SETDB1 not only associating with POAG, but also showed that they were expressing in the eye and particularly in the trabecular meshwork [71, 72]. Transforming growth factor-β (TGFβ) has the basic property of regulating and remodelling the extracellular matrix and hence is one of the candidate genes for glaucoma. TGFB1 –509C > T polymorphism is associated with POAG and therefore we looked at 104 patients with the disease but found no association of the SNP with VCDR, IOP and POAG [73]. VCDR is associated in glaucoma with ABCA1, ASAP1, ATOH7 and ELN gene polymorphisms [68]. GLIS1 (GLIS Family Zinc Finger 1 Kruppel-like transcription factor) variant rs941125 has shown to be associated with glaucoma in humans [74].

Though PXFG and POAG are the leading causes of blindness in glaucoma, PACG is one of the leading causes of blindness particularly in Asia and the blindness due to the latter (PACG) is 10 times more than that of POAG [75]. PLEKHA7, COL11A1, PCMTD1 and ST18 genes related SNPs located in chromosomes 11p15, 1p21 and 8q11.23 were first associated with PACG [76, 77, 78]. In major five Asian countries, a collaborative study was conducted in which 854 cases and 9608 controls (Singapore, Hong Kong, India, Malaysia and Vietnam) with replication studies on 1917 cases and 8943 controls (China, Singapore, India, Saudi Arabia and the UK, including that of the first author [GKM] team) GWAS was conducted to identify genetic factors’ associated with the PACG. In the GWAS, three SNPs were significantly associated with PACG in our collaborative cohort - rs11024102 in PLEKHA7 [Pleckstrin Homology Domain Containing A7] (per-allele odds ratio (OR) = 1.22; P = 5.33 × 10(−12)), rs3753841 in COL11A1 [Collagen Type XI Alpha 1 Chain] (per-allele OR = 1.20; P = 9.22 × 10(−10)) and rs1015213 located between PCMTD1 [Protein-L-Isoaspartate (D-Aspartate) O-Methyltransferase Domain Containing 1] and ST18 [ST18 C2H2C-Type Zinc Finger Transcription Factor] on chromosome 8q (per-allele OR = 1.50; P = 3.29 × 10(−9)) [76]. PLEKHA7 (Pleckstrin Homology Domain Containing, Family A Member 7) protein is require for zonule adherens biogenesis and maintenance, COL1A1 implicated in myopia and MMP9 have been also associated with ACG predisposing traits [79, 80]. COL11A1 (Collagen Type XI Alpha 1 Chain) protein may play a role in fibrillogenesis regulating the lateral growth of collagen II fibrils. PCMTD1 (Protein-L-Isoaspartate (D-Aspartate) O-Methyltransferase Domain Containing 1) protein is of the methyltransferase superfamily and ST18 (ST18 C2H2C-Type Zinc Finger Transcription Factor) protein inhibits basal transcription activity through target promoters. There are a myriad of players implicated in PACG as predisposing traits, (extensively reviewed by Ahram et al., Aboobakar and Wiggs) like MTHFR, MFRP, CHX10, HGF, RS; PO1, C3orf26, LAMA2, GJD2, ZNRF3, CD55, MIP, ALPPL2, ZC3H11B, PRSS56, ABCC5, MYOC, CYP1B1, eNOS, PCMTD1, ST18, HSP70, SPARC, CALCRL, EPDR1, CHAT, FERMT2, DPM2, FAM102A and NEB [77, 81]. A variety of anatomical, physiological, genetic and environmental factors individually or collectively result in PACG and hence, these associations reveal the larger etiopathogenesis network. There are many SNPs associated with PACG predisposing traits, however, the only gene so far identified which causes ACG is NNO1, which leads to nanophthalmos and hyperopia as well [77].

Primary pseudoexfoliation syndrome (PXFS) has fibrogranular extracellular debris in the anterior segment (besides systemic manifestations) which is made up of complex glycoprotein–proteoglycan that causes glaucoma in many but not all, with a preponderance in Scandinavian and Greek populations. In our cohort, we looked at LOXL1 [Lysyl Oxidase-Like Protein 1] gene exon 1 polymorphisms - allele G of rs1048661 (R141L) and allele G of rs3825942 (G135D), which are significantly associated with XFS in various populations [82]. About 52 XFS including those with glaucoma were screened for the variations and found that allele G of rs3825942 was significantly associated (p = 0.0001) and genotype GG (p = 0.000305) with XFS in our population, which was the first Asian study [83]. Pseudo-exfoliation glaucoma is caused by polymorphisms in the lysyl oxidase like 1 (LOXL1) gene in chromosome 15 with significant associations through GWAS in many populations across the world [81]. Pseudo-exfoliation syndrome, due to the deposition of extracellular fibrillar material (basement membrane, clusterin, elastic fibre contents, elastin, fibrillin-1, laminin, fibronectin, latent TGF-B proteins) crosslinking with LOXL1, hence systemically, it may be associated with cardiovascular diseases, cerebrovascular disorders, dementia like Alzheimer’s, pelvic organ prolapse and sensory neural deafness [84]. Non-coding variants in exons 1 and 2 of LOXL1 had conflicting reports [78]. CACN1A1 (Calcium Voltage-Gated Channel Subunit Alpha1 A) gene SNP variant was found to be significantly associated with ACG in the Japanese population which was validated in 17 other countries [78]. CACN1A1 helps calcium ion channel function and hence any dysregulation leads to the accumulation of XFS material on the trabecular meshwork. However, neurological disorders, familial hemiplegic migraine, epilepsy, cerebellar atrophy and episodic ataxia are associated with mutations in this gene. Another large 24 countries study significantly associated with POMP (proteasome maturation protein), TMEM136 (transmembrane protein 136), AGPAT1 (1-acylglyceroal-3phosphate O-acyltransfrase), RMBS3 (RNA binding motif single stranded interacting protein 3), SEMA6A (semaphorin 6A) and they were dysregulated [78]. In another Chinese study, The SNPs associated with the genes DENND1A (rs2479106), INSR (rs2059807), THADA (rs12478601), and TOX3 (rs4784165) [85].

Advertisement

5. Genomic mechanisms of glaucoma

The mechanisms of glaucoma is not understood clearly, however, increased IOP is significantly associated with structural (histopathological) and functional (physiological and molecular) distortions leading to neurodegeneration and glaucomatous modifications [86]. The mechanical physical strain and in addition collective stress effects (oxidative, reduced vascular flow, neurotrophic factors deprivation, metabolic, circulatory, immune, mitochondrial dysfunction, excitotoxicity, neuroinflammation, genetic susceptibility, vascular dysregulation) imposed on the lamina cribrosa, retinal ganglion cells and nearby optic nerve cells prevents the free flow of the axonal transport [86, 87]. Zukerman et al., in a review gave the list of genes associated with increased IOP, namely (in the same order)—ABCA1 (ATP-Binding Cassette, Sub-Family A (ABC1), Member 1), ABO (alpha 1–3-N-acetylgalactosaminyltransferase and alpha 1–3-galactosyltransferase), ADAMTS8 (ADAM Metallopeptidase With Thrombospondin Type 1 Motif 8), ADAMTS17 (ADAM Metallopeptidase With Thrombospondin Type 1 Motif 17), ADAMTS18-NUDT7,(ADAM Metallopeptidase With Thrombospondin Type 1 Motif 18- Nudix Hydrolase 7), AFAP1(Actin Filament Associated Protein), ANGPT1(Angiopoietin 1), ANTXR1(ANTXR Cell Adhesion Molecule 1), ARHGEF12 (Rho Guanine Nucleotide Exchange Factor 12), ARID5B (AT-Rich Interaction Domain 5B), ATXN2 (Ataxin 2), CAV1-CAV2(Caveolin 1- Caveolin 2), CDKN2B-AS1(CDKN2B Antisense RNA 1), CELF1 (CUGBP Elav-Like Family Member 1), CYP26A1-MYOF (Cytochrome P450 Family 26 Subfamily A Member 1- Myoferlin), FAM125B, (Family With Sequence Similarity 125, Member B) FNDC3B(Fibronectin Type III Domain Containing 3B), FOXC1 (Forkhead Box C1), FOXP1 (Forkhead Box P1), GAS7 (Growth Arrest Specific 7), GLCCI1-ICA1 (Glucocorticoid Induced 1- Islet Cell Autoantigen 1), GLIS3 (GLIS Family Zinc Finger 3), GMDS (GDP-Mannose 4,6-Dehydratase), HIVEP3 (HIVEP Zinc Finger 3), INCA1 (Inhibitor Of CDK, Cyclin A1 Interacting Protein 1), LMX1B (LIM Homeobox Transcription Factor 1 Beta), LOC171391, MADD (MAP Kinase Activating Death Domain), MIR548F3 (MicroRNA 548f-3), MYBPC3 (Myosin Binding Protein C3), NDUFS3 (NADH:Ubiquinone Oxidoreductase Core Subunit S3), NR1H3 (Nuclear Receptor Subfamily 1 Group H Member 3), PDDC1 (Parkinson disease 7 domain containing 1), PKHD1 (PKHD1 Ciliary IPT Domain Containing Fibrocystin/Polyductin), PTPRJ (Protein Tyrosine Phosphatase Receptor Type J), RAPSN (Receptor Associated Protein of the Synapse), RPLP2-PNPLA2 (Ribosomal Protein Lateral Stalk Subunit P2- Patatin Like Phospholipase Domain Containing 2), SIX1/SIX6 (SIX Homeobox 1/SIX Homeobox 6), SEPT9 (Septin 9), SEPT11 (Septin11), TFEC-TES (Transcription Factor EC- Testin LIM Domain Protein), TMCO1 (Transmembrane And Coiled-Coil Domains 1) and TXNRD2 (Thioredoxin Reductase 2) [2]. Majority of the genes’ mechanism to cause glaucoma is not understood, however, LMX1B (LIM homeodomain) alters anterior segment development and aqueous humour dynamics; MADD (MAP kinase activating death domain) performs through TNF-a-mediated microglial activation; NR1H3, a nuclear receptor, changes IOP through ABCA1 regulated aqueous humour dynamic alterations and SEPT9, a septin protein, acts through cytoskeletal alterations [2]. In the eye, genes could specifically act at certain parts, like trabecular meshwork (LMX1B, ABCA1), ciliary body (LMX1B), lamina cribrosa (ELN), superficial retinal nerve fibre layer (NR1H3, ABCA1, MADD, ASAP1, ATOH7) and prelaminar region (SEPT9) region [88]. LMX1B mutations have been associated with nail-patella syndrome (nail dysplasia, the patella is absent or is hypoplastic, chronic kidney disease) and a third of these patients develop glaucoma, due to increased IOP [89].

There are several genes significantly associated with CDR, namely, as cited alphabetically by Zukerman et al., − ABCA1 (ATP-Binding Cassette, Sub-Family A (ABC1), Member 1), ABG, ADAMTS8 (ADAM Metallopeptidase With Thrombospondin Type 1 Motif 8), ASAP1 (ArfGAP With SH3 Domain, Ankyrin Repeat And PH Domain 1), ASB7 (Ankyrin Repeat And SOCS Box Containing 7), ATOH7 (Atonal BHLH Transcription Factor 7), ATOH7-PBLD (Atonal BHLH Transcription Factor 7- Phenazine Biosynthesis Like Protein Domain Containing), BMP2 (Bone Morphogenetic Protein 2), CARD10 (Caspase Recruitment Domain Family Member 10), CDC7-TGFBR3 (Cell Division Cycle 7- Transforming Growth Factor Beta Receptor 3), CDKN2B (Cyclin Dependent Kinase Inhibitor 2B), CDKN2B-CDKN2BAS (Cyclin Dependent Kinase Inhibitor 2B-CDKN2B Antisense RNA 1), CHEK2 (Checkpoint Kinase 2), COL8A1 (Collagen Type VIII Alpha 1 Chain), CRISPLD1 (Cysteine Rich Secretory Protein LCCL Domain Containing 1), DCLK1 (Doublecortin Like Kinase 1), DGKB (Diacylglycerol Kinase Beta), DUSP1 (Dual Specificity Phosphatase 1), ELN (Elastin), ENO4 (Enolase 4), EXOC2 (Exocyst Complex Component 2), F5 (Coagulation Factor V), FAM101A (Family With Sequence Similarity 101, Member A), GAS7 (Growth Arrest Specific 7), HSF2 (Heat Shock Transcription Factor 2), PDZD2 (PDZ Domain Containing 2), PLCE1 (Phospholipase C Epsilon 1), PSCA (Prostate Stem Cell Antigen), RARB (Retinoic Acid Receptor Beta), RERE (Arginine-Glutamic Acid Dipeptide Repeats), RPAP3 (RNA Polymerase II Associated Protein 3), RPE65 (Retinoid Isomerohydrolase RPE65), RREB1 (Ras Responsive Element Binding Protein 1), SALL1 (Spalt Like Transcription Factor 1), SCYL1 (SCY1 Like Pseudokinase 1), SIX1 (SIX Homeobox 1), SIX6 (SIX Homeobox 6), SSSCA1 (Sjogren’S Syndrome/Scleroderma Autoantigen 1), TMTC2 (Transmembrane O-Mannosyltransferase Targeting Cadherins 2), and VCAN (Versican) [90]. Some of them have been linked with both CDR and IOP - ABCA1, ABG, AFAP1, CAV1, GAS7 and LMX1B [2, 91]. RPE65 gene mutations result in Leber congenital amaurosis and early childhood onset retinitis pigmentosa [92]. ABCA1 is associated with cholesterol metabolism and liver function and is associated with retinal ganglion cell death and normal physiology [91]. ELN modifies the normal activity of elastin resulting in optic nerve head degeneration; ASAP1 is associated with giant cell medicated retinal ganglion cell loss and ATOH7 is connected with Muller cell differentiation and retinal ganglion cell genesis [90]. The degenerative patterns are seen in different structures of the RGC—soma atrophy, nuclear shrinkage axonic insult, and deteriorating changes in the synapses and dendrites, finally extending to the amacrine and bipolar cells [87]. Adding to the complexity, a transcriptome wide association studies identified SIX6 and CDKN2A/B to be associated with POAG and these are also linked to cardiovascular diseases and cancer [93]. The mitogen activated protein kinase p38 and Jun N terminal kinases are activated through several signalling pathways which initiate the degeneration of the soma of the RGC [94]. Subsequently, activation of the apoptotic pathway is triggered and the BCL2 gene family BAX is prompted in monkeys, rabbits and humans as well [95, 96].

Advertisement

6. Neuroprotection & neurodegeneration genomics in glaucoma

Glaucoma is nowadays considered to be a chronic neurodegenerative disorder which has decreased sensitivity to colour and contrast, blurry vision and reducing the field of vision with nil signs or symptoms [45]. The transcription factors, transporters, glycosylation proteins, and mutations will result in loss of function, low-risk variants gene expression modifications due to RNA splicing and transcription activities. Epigenetic activities like DNA methylation, histone body acetylation, deacetylation, structural chromatin modification and transcription. Micro RNAs miR24, miR29, miR204, miR146a [45]. In mice with optic nerve crush and glaucomatous damage could be rescued with miR-194 and miR-644-2 inhibitors provided neuroprotection and miR-181a and miR-181d-5p mimics showed neuritogenesis in retinal ganglion cells [97].

Optic nerve head is the primary critical site of degeneration in glaucoma. Axonal deprivation of neurotrophins like brain derived neurotrophic factors and mitochondrial dysfunction leads to axon transport failure [98]. There are other stakeholders in axonal degeneration of RGCs, like reduced blood flow, extracellular matrix remodelling, oxidative stress and reactive gliosis [99]. The Rho/ROCK signalling pathway prevents central nervous system regeneration through transducing inhibitory signals and is a good target for intervention in axon regeneration in glaucoma [100]. P13K/Akt pathway facilitates axonal growth and regeneration by converting PIP2 (phosphatidylinositol bisphosphate) to PIP3 (phosphatidylinositol trisphosphate) which in turn activates protein kinase Akt. This action results in phosphorylation and activation of mTOR (rapamycin), which promotes protein synthesis, motility, cell growth and survival [101]. Jak/STAT is involved in axonal regeneration by the binding of cytokines to the extracellular receptors associated with protein kinase JAK, which activates and phosphorylates the STATs. Axon regeneration is inhibited by the suppressor of cytokine signalling (SOCS) suppressing the Jak/STAT signalling [101]. Interestingly, in an immunoreactive male Lewis rats for S100B protein (the antibody which is found in high titre in glaucoma patients) showed 43 proteins were dysregulated in the retina, out of which alpha-2 macroglobulin increase was significantly associated with heat shock protein 60, showcasing the role of immunological factors in glaucoma [102].

Glaucomatous pressure leads to the progressive death of the retinal ganglion cells (RGCs), degeneration of the optic nerve and loss of peripheral vision, though normal tension glaucoma happens with the same pathological mechanism, questioning the role of intraocular pressure in the process. The trabecular meshwork is the seat of the pathology with a number of influencing factors like ageing, genetics, mechanical and oxidative stress, all collectively inhibiting the neurotrophic molecules nourishing the RGCs.

Neuroprotection of the retinal ganglion cells is critical for cell survival since several signalling pathways play the role, like JAK/STAT, MAPK, TrkA, TrkB and clinical trials with CNTF (ciliary neurotrophic factor and NGF [nerve growth factor] are in vogue [103]. CNTF is a neuropoietic cytokine belonging to IL6, which binds to the receptor of gp130 to activate JAK/STAT and MAPK to neuroprotect the RGCs [103]. NGF is secreted by nerve tissue (neurons, oligodendrocytes, Schwann cells), immune cells (T cells, mast cells, macrophages), skin cells (fibroblasts, keratinocytes, melanocytes) and smooth cells, which regulate apoptosis, neuronal plasticity, neurogenesis and neuroinflammation [103]. BDNF (brain-derived nerve growth factor), VEGF (vascular endothelial growth factor), PEDG (pigment epithelium-derived factor), GDNF (glial cell line derived neurotrophic factor) and Norrin are some of the other RGC neuroprotective proteins [103].

Epithelial cells, glial cells, leukocytes and neurons produce various neuroprotective factors like brain-derived neurotrophic factor, ciliary neurotrophic factor, glial cell line derived neurotrophic factor, nerve growth factor, norrin, pigment epithelium-derived factor, vascular endothelial growth factor and each in an exceptional way prevent RGC damage which is triggered by the ischaemic neuropathy, glaucoma, ocular hypertension and oxygen-induced retinopathy and the survival is achieved by interventional strategy through activating a variety of signalling pathways like JAK/STAT, MAPK, TrkA and TrKB [103].

The cell and tissue stakeholders in glaucoma are trabecular meshwork, retinal ganglion cell layer, retinal nerve fibre layer, cells in the optic nerve head (lamina cribrosa, optic nerve head astrocytes) and peripapillary sclera around the optic nerve head [104]. These components react to biomechanical stress like compression and stretching and the cell structures that respond are the cell membrane, cytoskeleton (actin microfilaments and tubules), extracellular matrix and nucleus. Gene expression, hence, in these cells are altered with copious TGFbeta2 synthesis in glaucoma models [104].

6.1 Biomarker genomics in glaucoma

Protein biomarkers have been identified in various parts of the eye structure associated with glaucoma, as explained in the review by Cueto et al. [1]. However, caution has to be adopted while interpreting the protein biomarker studies, due to the nature of these studies where different clinical and laboratory methodologies with variable sensitivity and specificity techniques and equipments were used, the sample sizes were too small, many studies are not validated, there are conflicting reports of dysregulation and there is poor consensus, no data between, the aqueous humour, tears, serum and vitreous samples. However, overexpression of the biomarkers could become neurotoxic and down-regulation and lack of or less expression of neuroprotectors will lead to degeneration of the retinal ganglion cells via the TrkA receptor pathway. Biomarkers could provide early screening and detection of glaucoma in the target population, diagnosis and prognostication. The biomarkers upstream or downstream could be novel targets for therapeutic interventions and visual stability or recovery. Accumulation of biomarkers will distort the blood aqueous barrier due to the inflammation and dysregulation of the extracellular matrix tissue physiology. Similarly, the biomarkers will intervene in the autonomic regulation of the sympathetic system affecting the ciliary body and trabecular meshwork physiological architecture. To date, over 450 biomarkers have been identified which have never been validated across large sample size patients and controls, not across different populations in the world and have not entered the arena of clinical practice, keeping the research door wide open.

Biomarkers have been identified in aqueous humour, optic nerve, retina, trabecular meshwork, tears, vitreous body, serum and blood. Besides, there are biomarkers related to apoptosis, inflammation, oxidative stress, extracellular matrix, immune response, neuroprotection, and neurodegeneration. A fairly extensive list of the biomarkers in glaucoma is provided in Table 1, please refer to Cueto et al., for detailed information [1].

Serial numberBiomarkers typeBiomarkers
1Inflammatory biomarkersIncreased: TGFB2, CD44, erythropoietin, TNFA, IL8, serum amyloid A, CXCL13, CXCL16, CCL13, CCL15, CCL22, CCL24, IL-4, IL-16 (PXFG); autotoxin, Growth differentiation protein 15 and endothelin, Proatrial natriuretic peptide (regulates vascular/neural integrity of adult retina), IL-5, IL-12, IL-15, interferon gamma, fibroblast growth factor, vascular endothelial growth
Decreased: Secreted frizzled related protein-1, klotho (ageing protein),
2.Oxidative stress related biomarkersIncreased: superoxide dismutase, glutathione peroxidase, malondialdehyde, nitric oxide synthase, carbonyl, hydrogen peroxide, advanced glycation end products.
Decreased: Catalase, vitamins C/E
3.Extracellular matrix related biomarkersIncreased: Fibronectin; clusterin; periostin
Decreased: Hyaluronic acid, fibulin-7,
Variably expressed: Connective tissue growth factor, gelatinase.
Under regulated: Cystatin C, osteopontin,
4.Immune-response-, neurodegeneration-, and apoptosis-related markersIncreased: Heat shock protein-70, vimentin; heat shock protein-27, transthyretin; prostaglandin H2 D-isomerase, caspase 14 precursor, CysC, albumin precursor, transferrin; apolipoprotein A4, ALB, antithrombin 3 (SERPINC1), CD14, CD59, complement factor D, APOA4, chromogranin A, MYB, TIMP1, microfibril-associated glycoprotein 4, agrin, and apolipoprotein C-III, Ig j chain C region, inter-a-trypsin inhibitor heavy chain 4, isocitrate dehydrogenase (NAD) subunit α, ALB, CysC, TIMP2, A2M, PGTDS, NPP2, apolipoprotein A1, APOC3, apolipoprotein E, transthyretin, and α2-macroglobulin, vitronectin, complement factors (C3a, C5b-9),
Decreased: α-enolase (ENO1), actin, and glyceraldehyde-3-phosphate dehydrogenase (POAG, PEXG), transthyretin, prostaglandin H2D isomerase, opticin, interphotoreceptor retinoid-binding protein, apolipoprotein D, SOD1,
5.Metabolite based biomarkersIncreased: Homocysteine, diadenosine tetraphosphate, MDA, creatinine, carnitines, aminoacids (glutamine, glycine, alanine, leucine, isoleucine, hydroxyproline, acetylornithine), several phosphatidylcholines, lysophosphatidylcholines, sphingomyelin, glycine (significantly different), pelargonic acid and galactose 1, glucose-1 phosphate, sorbitol, spermidine 2, betaine, taurine, glutamate,
Decreased: Adenosine triphosphate/ Adenosine diphosphate, taurine, spermine
6.Lipid metabolismIncreased: palmitoleic acid, gamma-linolenic acid, arachidonic acid, adrenic acid, hydroxylinoleate, hydroxyarachidonate isomers
Decreased: eicosapentaenoic fatty acid, DHA, total ω3 long-chain polyunsaturated fatty acid

Table 1.

Dysregulation of gene expression and biomarkers in primary open angle glaucoma, primary closed-angle glaucoma, primary congenital glaucoma, pseudo-exfoliation glaucoma and neovascular glaucoma are summarised. Please refer to Ceutu et al., for further details [1].

6.2 Recent advances of genomic interventional strategies & glaucoma

Gene therapy in glaucoma is promising and is tackled by neuroprotection of the focusing on prevention of neuronal cell soma and axon loss. Another method is of optic nerve axonic regeneration [87]. In neuroprotection gene therapy, mostly in animal studies, what is addressed are overexpressing of growth and neurotrophic factors (brain-derived neurotrophic factor, fibroblast growth factor, ciliary neurotrophic factor), antiapoptotic factors (BAG1, Bcl-X, BIRC4/XIAP), transcription factors (ATF3, Brn3b, CREB, NMDA, KLF7), oxidative stress components (catalase, NRF2, SOD2), Rho/ROCK pathway (exoenzyme C3, RhoA, ROCK2), mitochondrial targets (NMNAT1, DBA2J, OPA1) and other targets (ABCA1, MCT2, Hsp70, MEK1, ULK1, miRNAs) [87]. On the axon regeneration gene therapy, what is targeted are either by overexpression or silencing in the optic nerve, optic chiasma, optic tract - PI3K/Akt pathway (PTEN, P13K, cRHEB, S6K1, GSK3, eIF2B, FGF2, IGF1, neuretin), Jak/STAT pathway (CNTF, IL6, IL22, STAT3, SOCS4, Pim1), Rho/ROCK pathway (RhoA, ROCK2, LIMK-1, LOTUS, PirB), transcription factors (KLF9, c-myc, KLF4, p53, SOX11) and other targets (many including, Lin28, HDAC5, melanopsin, TIMP2, PRPH). The gene and molecules list are selective and not exhaustive. These therapies could also be given in a combinatorial manner. Many of these molecules are awaiting the approval of the FDA, USA for clinical trials [87].

Advertisement

7. Experimental bioinformatics analysis

We used a bioinformatic analysis to arrive at the exhaustive list of genes or genetic factors associated with glaucoma. Genes and variants associated with different types of glaucoma were mined by using the DisGeNET Cytoscape App (version 7.0) [105]. The DisGeNET database, retrieves gene-disease and variant-disease associations from curated databases. Analysis was performed for “Gene Disease Networks” and “Variant Disease Network”, by selecting “curated” as source and “Eye diseases” as disease class and “Glaucoma” as disease. The plethora list of genes and genetic factors are provided according to the type of glaucoma in Tables 29.

GeneGene_Full_NameProtein_Class
CYP1B1Cytochrome P450 family 1 subfamily B member 1Enzyme
LTBP2Latent transforming growth factor beta binding protein 2Calcium-binding protein
MYOCMyocilinCellular structure

Table 2.

Genes associated with Juvenile open-angle glaucoma.

GeneVariantChrPositionConsequenceAllelesClass
CYP1B1rs104893629238071087missense variantT/Asnv

Table 3.

Variants associated with Juvenile open-angle glaucoma.

GeneGene Full NameProtein Class
ADAMTSL1ADAMTS like 1Enzyme
ADRB2Adrenoceptor beta 2G-protein coupled receptor
ANGPT1Angiopoietin 1Signaling
ARSDArylsulfatase DEnzyme
COL1A1Collagen type I alpha 1 chain
CYP1B1Cytochrome P450 family 1 subfamily B member 1Enzyme
CYP2B6Cytochrome P450 family 2 subfamily B member 6
FOXC1Forkhead box C1Transcription factor
GLC3BGlaucoma 3, primary infantile, B
GLC3CGlaucoma 3, primary congenital, C
HTC2Hypertrichosis 2 (generalized, congenital)
KIF1BKinesin family member 1BCellular structure
LOC110599580CYP1B1 promoter
LOXL1Lysyl oxidase like 1
LTBP2Latent transforming growth factor beta binding protein 2Calcium-binding protein
MFN2Mitofusin 2Enzyme
MYOCMyocilinCellular structure
PGCProgastricsinEnzyme
PLXNA2Plexin A2
SH3PXD2BSH3 and PX domains 2B
SLC4A4Solute carrier family 4 member 4Transporter
STATHStatherin
TEKTEK receptor tyrosine kinaseKinase
TYRTyrosinaseEnzyme

Table 4.

Genes associated with primary congenital glaucoma.

Literature suggests that the inheritance of PCG includes an autosomal-recessive and sex-associated element with variable penetrance. Over 150 variants identified in CYP1B1 gene are responsible for the of PCG. Various studies showed the genes (CYP1B1, LTBP2, MYOC, COL1A1, FOXC1, ANGTP1, TEK) associated with the pathogenesis of PCG.

GeneVariantChrPositionConsequenceAllelesClass
ADRB2rs10427145148826910Stop gainedG/C;Tsnv
ADRB2rs18008885148827322Missense variantC/Tsnv
COL1A1rs726453181750197057Stop gainedG/Asnv
COL1A1rs726516581750190861Missense variantC/Tsnv
CYP1B1rs79204362238071251Missense variantC/Tsnv
CYP1B1rs104893622238071234Missense variantC/Tsnv
CYP1B1rs1800440238070996Missense variantT/C;Gsnv
CYP1B1rs55989760238071195Missense variantC/G;Tsnv
CYP1B1rs56010818238071185Missense variantC/Tsnv
CYP1B1rs72549379238071264Missense variantC/Tsnv
CYP1B1;CYP1B1-AS1rs28936700238075207Missense variantC/G;Tsnv
CYP1B1;CYP1B1-AS1rs104893623238075219Stop gainedC/Tsnv
CYP1B1;CYP1B1-AS1rs1272655298238074527Missense variantC/G;Tsnv
CYP1B1;CYP1B1-AS1rs2567206238076389Non coding transcript exon variantG/Asnv
CYP1B1;CYP1B1-AS1rs72481807238074872Stop gainedC/A;Tsnv
CYP1B1;CYP1B1-AS1rs9282671238075148Missense variantA/Tsnv
CYP1B1-AS1;CYP1B1rs57865060238074704Missense variantC/Tsnv
CYP1B1-AS1;CYP1B1rs72549387238075218Stop gainedC/G;Tsnv
FN1rs12779892972215428270Stop gainedG/Asnv
LTBP2rs1219183551474555629Stop gainedG/A;Tsnv
LTBP2rs37427931474603790Intron variantG/Csnv
LTBP2rs617380251474552299Synonymous variantC/Tsnv
MYOCrs743153391171652468Missense variantC/Asnv
MYOC;MYOCOSrs7528291381171638607Frameshift variantTC/-delins
PAX6rs1219079171131794079Stop gainedG/Asnv

Table 5.

Variants associated with primary congenital glaucoma.

GeneGene_Full_NameProtein_Class
ABCA1ATP binding cassette subfamily A member 1Transporter
ABCB1ATP binding cassette subfamily B member 1Transporter
ABCC4ATP binding cassette subfamily C member 4Transporter
ABOABO, alpha 1-3-N-acetylgalactosaminyltransferase and alpha 1-3-galactosyltransferaseEnzyme
ACEAngiotensin I converting enzymeEnzyme
ACOT7Acyl-CoA thioesterase 7Enzyme
ACTBActin betaCellular structure
ACTBL2Actin beta like 2
ACTG1Actin gamma 1Cellular structure
ACTG2Actin gamma 2, smooth muscleCellular structure
ADAMTS10ADAM metallopeptidase with thrombospondin type 1 motif 10Enzyme
ADAMTS17ADAM metallopeptidase with thrombospondin type 1 motif 17Enzyme
ADAMTSL3ADAMTS like 3Enzyme
ADRB2Adrenoceptor beta 2G-protein coupled receptor
AFAP1Actin filament associated protein 1
AGBL2ATP/GTP binding protein like 2Enzyme
AGERAdvanced glycosylation end-product specific receptorReceptor
AKT1AKT serine/threonine kinase 1Kinase
ALBAlbuminTransporter
ANGPT2Angiopoietin 2Signaling
ANGPTL7Angiopoietin like 7Signaling
ANXA5Annexin A5
APBB2Amyloid beta precursor protein binding family B member 2
APEX1Apurinic/apyrimidinic endodeoxyribonuclease 1
APOC3Apolipoprotein C3
APOEApolipoprotein E
APPAmyloid beta precursor proteinEnzyme modulator
AQP1Aquaporin 1 (Colton blood group)Ion channel
ARHGEF12Rho guanine nucleotide exchange factor 12
ARHGEF7Rho guanine nucleotide exchange factor 7
ARSDArylsulfatase DEnzyme
ASB10Ankyrin repeat and SOCS box containing 10
ASCC1Activating signal cointegrator 1 complex subunit 1
ASCC2Activating signal cointegrator 1 complex subunit 2
ATOH7Atonal bHLH transcription factor 7Enzyme
ATP10AATPase phospholipid transporting 10A (putative)Transporter
ATXN2ataxin 2Nucleic acid binding
AXLAXL receptor tyrosine kinaseKinase
B4GALT3Beta-1,4-galactosyltransferase 3Enzyme
BAK1BCL2 antagonist/killer 1Signaling
BDNFBrain derived neurotrophic factorSignaling
BIRC6Baculoviral IAP repeat containing 6
BMP4Bone morphogenetic protein 4Signaling
BRCA1BRCA1 DNA repair associatedEnzyme
C1QBPComplement C1q binding protein
C3Complement C3Enzyme modulator
CACNA1CCalcium voltage-gated channel subunit alpha1 CIon channel
CACNA2D1Calcium voltage-gated channel auxiliary subunit alpha2delta 1Ion channel
CALCACalcitonin related polypeptide alphaSignaling
CALCRLCalcitonin receptor like receptorG-protein coupled receptor
CARD10Caspase recruitment domain family member 10
CATCatalaseEnzyme
CAV1caveolin 1Enzyme modulator
CAV2caveolin 2Enzyme modulator
CCHCR1Coiled-coil alpha-helical rod protein 1
CCL16C-C motif chemokine ligand 16Signaling
CCL2C-C motif chemokine ligand 2Signaling
CCL4C-C motif chemokine ligand 4Signaling
CCL4L1C-C motif chemokine ligand 4 like 1
CCL4L2C-C motif chemokine ligand 4 like 2
CCN2Cellular communication network factor 2Signaling
CD40CD40 molecule
CDC7Cell division cycle 7Kinase
CDH1Cadherin 1
CDH5Cadherin 5
CDK9Cyclin dependent kinase 9Kinase
CDKN1ACyclin dependent kinase inhibitor 1A
CDKN2ACyclin dependent kinase inhibitor 2A
CDKN2BCyclin dependent kinase inhibitor 2B
CDKN2B-AS1CDKN2B antisense RNA 1
CDX2Caudal type homeobox 2Transcription factor
CHDHCholine dehydrogenaseEnzyme
CIAO3Cytosolic iron-sulfur assembly component 3Enzyme
CLCN2Chloride voltage-gated channel 2Ion channel
CLUClusterin
CNTFCiliary neurotrophic factor
CNTN4Contactin 4Receptor
CNTNAP4Contactin associated protein family member 4
COCHCochlinReceptor
COL11A1Collagen type XI alpha 1 chain
COL15A1Collagen type XV alpha 1 chain
COL18A1Collagen type XVIII alpha 1 chain
COL1A1Collagen type I alpha 1 chain
COL5A1Collagen type V alpha 1 chain
COL5A2Collagen type V alpha 2 chain
COL8A1Collagen type VIII alpha 1 chainExtracellular structure
COL8A2Collagen type VIII alpha 2 chainExtracellular structure
COX1Cytochrome c oxidase subunit IEnzyme
COX2Cytochrome c oxidase subunit IIEnzyme
CRISP2Cysteine rich secretory protein 2Immune response
CRYABCrystallin alpha B
CST3Cystatin C
CTSDCathepsin DEnzyme
CUX1Cut like homeobox 1Transcription factor
CXCL5C-X-C motif chemokine ligand 5Signaling
CXCR3C-X-C motif chemokine receptor 3G-protein coupled receptor
CYP1A1Cytochrome p450 family 1 subfamily a member 1Enzyme
CYP1B1Cytochrome p450 family 1 subfamily b member 1Enzyme
CYP27A1Cytochrome p450 family 27 subfamily a member 1Enzyme
CYP2C19Cytochrome p450 family 2 subfamily c member 19
CYP2D6Cytochrome p450 family 2 subfamily d member 6
CYP46A1Cytochrome p450 family 46 subfamily a member 1Enzyme
DBN1Drebrin 1Cellular structure
DCLK1Doublecortin like kinase 1Kinase
DCNDecorin
DDIT3DNA damage inducible transcript 3
DDX20DEAD-box helicase 20
DDX3XDEAD-box helicase 3 X-linked
DLG2Discs large MAGUK scaffold protein 2Receptor
DNASE1L3Deoxyribonuclease 1 like 3
EBF1EBF transcription factor 1
EDN1Endothelin 1Signaling
EDNRAEndothelin receptor type AG-protein coupled receptor
EFEMP1EGF containing fibulin extracellular matrix protein 1Extracellular structure
EGFREpidermal growth factor receptorKinase
EGR1Early growth response 1Nucleic acid binding
EIF2DEukaryotic translation initiation factor 2DReceptor
ELNElastin
ELOVL5ELOVL fatty acid elongase 5Enzyme
ESR1Estrogen receptor 1Nuclear receptor
ESR2Estrogen receptor 2Nuclear receptor
FASTKD1FAST kinase domains 1
FBLN1Fibulin 1
FBLN5Fibulin 5Calcium-binding protein
FBLN7Fibulin 7
FBN1Fibrillin 1Calcium-binding protein
FHL5Four and a half LIM domains 5Transcription factor
FLNBFilamin B
FLOT1Flotillin 1
FN1Fibronectin 1Signaling
FNDC3BFibronectin type III domain containing 3B
FOXC1Forkhead box C1Transcription factor
FUT7Fucosyltransferase 7Enzyme
FZR1Fizzy and cell division cycle 20 related 1Enzyme modulator
GALCGalactosylceramidase
GAS1Growth arrest specific 1
GAS7Growth arrest specific 7
GDF15Growth differentiation factor 15Signaling
GJA1Gap junction protein alpha 1Cell-cell junction
GLB1Galactosidase beta 1Enzyme
GLC1BGlaucoma 1, open angle, B (adult-onset)
GLC1CGlaucoma 1, open angle, C
GLC1DGlaucoma 1, open angle, D (adult-onset)
GLC1HGlaucoma 1, open angle, H (adult-onset)
GLC1JGlaucoma 1, open angle, J (juvenile-onset)
GLC1KGlaucoma 1, open angle, K (juvenile-onset)
GLC1NGlaucoma 1, open angle, N (juvenile-onset)
GLC3BGlaucoma 3, primary infantile, B
GLCCI1Glucocorticoid induced 1
GMDSGDP-mannose 4,6-dehydrataseEnzyme
GRIN2BGlutamate ionotropic receptor NMDA type subunit 2BIon channel
GSTK1Glutathione S-transferase kappa 1
GSTM1Glutathione S-transferase mu 1
GSTM2Glutathione S-transferase mu 2
GSTP1Glutathione S-transferase pi 1
GSTT1Glutathione S-transferase theta 1
GUCY1A1Guanylate cyclase 1 soluble subunit alpha 1
H3P40H3 histone pseudogene 40
HAS2Hyaluronan synthase 2
HDAC6Histone deacetylase 6Epigenetic regulator
HES1Hes family bHLH transcription factor 1Transcription factor
HEYLHes related family bHLH transcription factor with YRPW motif likeTranscription factor
HK2Hexokinase 2Kinase
HLA-AMajor histocompatibility complex, class I, A
HLA-DQB1Major histocompatibility complex, class II, DQ beta 1Immune response
HLA-DRB1Major histocompatibility complex, class II, DR beta 1Immune response
HPGDSHematopoietic prostaglandin D synthase
HSPA14Heat shock protein family A (Hsp70) member 14
HSPA1AHeat shock protein family A (Hsp70) member 1A
HSPA1BHeat shock protein family A (Hsp70) member 1B
HSPA4Heat shock protein family A (Hsp70) member 4
HSPA5Heat shock protein family A (Hsp70) member 5
HSPB1Heat shock protein family B (small) member 1
HSPB2Heat shock protein family B (small) member 2
HSPB3Heat shock protein family B (small) member 3
HSPD1Heat shock protein family D (Hsp60) member 1
HTC2Hypertrichosis 2 (generalized, congenital)
HYAL3Hyaluronidase 3 hyaluronidase 3Enzyme
ICA1Islet cell autoantigen 1
IDH3AIsocitrate dehydrogenase (NAD(+)) 3 catalytic subunit alphaEnzyme
IFNGInterferon gamma
IGF2Insulin like growth factor 2
IGFALSInsulin like growth factor binding protein acid labile subunitReceptor
IGKCImmunoglobulin kappa constant
IL10Interleukin 10
IL17BInterleukin 17B
IL1AInterleukin 1 alpha
IL1BInterleukin 1 beta
IL1RNInterleukin 1 receptor antagonist
IL2Interleukin 2
IL20Interleukin 20
IL20RBInterleukin 20 receptor subunit betaReceptor
IL2RAInterleukin 2 receptor subunit alphaReceptor
IL6Interleukin 6
IL7Interleukin 7
IL9Interleukin 9
ISG20Interferon stimulated exonuclease gene 20
ITGA5Integrin subunit alpha 5
ITGAVIntegrin subunit alpha V
ITIH4Inter-alpha-trypsin inhibitor heavy chain 4Enzyme modulator
ITPR3Inositol 1,4,5-trisphosphate receptor type 3Ion channel
KDRKinase insert domain receptorKinase
LDLRLow density lipoprotein receptor
LGALS14Galectin 14Signaling
LGTNLigatin
LHCGRLuteinizing hormone/choriogonadotropin receptorG-protein coupled receptor
LINC02605Long intergenic non-protein coding RNA 2605
LMX1BLIM homeobox transcription factor 1 betaNucleic acid binding
LOC110599580CYP1B1 promoter
LOXL1Lysyl oxidase like 1
LOXL2Lysyl oxidase like 2
LTBP2Latent transforming growth factor beta binding protein 2Calcium-binding protein
MAP3K1Mitogen-activated protein kinase kinase kinase 1Kinase
MAP3K8Mitogen-activated protein kinase kinase kinase 8Kinase
MARCHF8Membrane associated ring-CH-type finger 8
MARCHF9Membrane associated ring-CH-type finger 9
MBL2Mannose binding lectin 2Receptor
MBPMyelin basic protein
MFGE8Milk fat globule-EGF factor 8 proteinEnzyme
MINDY4MINDY lysine 48 deubiquitinase 4
MIR182microRNA 182
MIR210microRNA 210
MIR302DmicroRNA 302d
MIR34BmicroRNA 34b
MIR630microRNA 630
MLXIPMLX interacting protein
MMP1Matrix metallopeptidase 1Enzyme
MMP12Matrix metallopeptidase 12Enzyme
MMP2Matrix metallopeptidase 2Enzyme
MMP3Matrix metallopeptidase 3Enzyme
MMP9Matrix metallopeptidase 9Enzyme
MMRN1Multimerin 1
MPOMyeloperoxidaseEnzyme
MPP7Membrane palmitoylated protein 7Enzyme
MT1AMetallothionein 1A
MT1BMetallothionein 1B
MT1EMetallothionein 1E
MT1FMetallothionein 1F
MT1GMetallothionein 1G
MT1HMetallothionein 1H
MT1IPMetallothionein 1I, pseudogene
MT1JPMetallothionein 1J, pseudogene
MT1LMetallothionein 1L, pseudogene
MT1MMetallothionein 1M
MT1XMetallothionein 1X
MTCO2P12MT-CO2 pseudogene 12
MTHFRMethylenetetrahydrofolate reductase
MTNR1AMelatonin receptor 1AG-protein coupled receptor
MUTYHmutY DNA glycosylaseEnzyme
MVB12BMultivesicular body subunit 12B
MYLIPMyosin regulatory light chain interacting protein
MYOCMyocilinCellular structure
MYOCOSMyocilin opposite strand
MZB1Marginal zone B and B1 cell specific protein
NANOS2Nanos C2HC-type zinc finger 2
ND2MTND2
NFKB1Nuclear factor kappa B subunit 1Transcription factor
NFKB2Nuclear factor kappa B subunit 2Transcription factor
NOS2Nitric oxide synthase 2
NOS3Nitric oxide synthase 3
NPPANatriuretic peptide A
NPPCNatriuretic peptide CSignaling
NR3C1Nuclear receptor subfamily 3 group C member 1Nuclear receptor
NTF4Neurotrophin 4Signaling
NTMNeurotrimin
NXF1Nuclear RNA export factor 1Nucleic acid binding
OAS32'-5'-oligoadenylate synthetase 3Enzyme
OGG18-oxoguanine DNA glycosylase
OGNOsteoglycin
OPA1OPA1 mitochondrial dynamin like GTPaseEnzyme modulator
OPTCOpticinReceptor
OPTNOptineurin
PADI2Peptidyl arginine deiminase 2
PAHPhenylalanine hydroxylase
PARP1Poly(ADP-ribose) polymerase 1
PCOLCE2Procollagen C-endopeptidase enhancer 2
PDE5APhosphodiesterase 5A
PDIA5Protein disulfide isomerase family A member 5
PEX5Peroxisomal biogenesis factor 5Transporter
PITX2Paired like homeodomain 2
PKHD1PKHD1 ciliary IPT domain containing fibrocystin/polyductin
PLA2G4APhospholipase A2 group IVAEnzyme
PLB1Phospholipase B1
PLGPlasminogenEnzyme
PLXDC2Plexin domain containing 2
PLXNA2Plexin A2
PMELPremelanosome proteinSignaling
POTEKPPOTE ankyrin domain family member K, pseudogene
POTEMPOTE ankyrin domain family member M
PPIDPeptidylprolyl isomerase D
PPIFPeptidylprolyl isomerase F
PRDM5PR/SET domain 5
PRKAA1Protein kinase AMP-activated catalytic subunit alpha 1Kinase
PRNPPrion protein
PRPF8Pre-mRNA processing factor 8Nucleic acid binding
PRSPrieto X-linked mental retardation syndrome
PRSS55Serine protease 55Enzyme
PSDPleckstrin and Sec7 domain containing
PTENPhosphatase and tensin homologEnzyme
PTGFRProstaglandin F receptorG-protein coupled receptor
PTGS1Prostaglandin-endoperoxide synthase 1Enzyme
PTGS2Prostaglandin-endoperoxide synthase 2Enzyme
PTPRJProtein tyrosine phosphatase receptor type JEnzyme
RAMP2Receptor activity modifying protein 2Receptor
RANRAN, member RAS oncogene familyEnzyme modulator
RBP1Retinol binding protein 1
RHOAras homolog family member AEnzyme modulator
RHODras homolog family member DEnzyme modulator
RNR2l-rRNA
ROCK1Rho associated coiled-coil containing protein kinase 1Kinase
ROCK2Rho associated coiled-coil containing protein kinase 2Kinase
ROS1ROS proto-oncogene 1, receptor tyrosine kinaseKinase
RPGRIP1RPGR interacting protein 1Enzyme modulator
RPN2Ribophorin IIEnzyme
RTCARNA 3'-terminal phosphate cyclaseEnzyme
SART3Spliceosome associated factor 3, U4/U6 recycling proteinNucleic acid binding
SCGB1A1Secretoglobin family 1A member 1Signaling
SEC14L2SEC14 like lipid binding 2
SELENBP1Selenium binding protein 1Immune response
SEMA6ASemaphorin 6ASignaling
SERPINA3Serpin family A member 3Enzyme modulator
SERPINE1Serpin family E member 1Enzyme modulator
SFRP1Secreted frizzled related protein 1
SH3PXD2BSH3 and PX domains 2B
SHBGSex hormone binding globulin
SIRT1Sirtuin 1Epigenetic regulator
SIX1SIX homeobox 1Transcription factor
SIX6SIX homeobox 6Transcription factor
SLC23A1Solute carrier family 23 member 1Transporter
SLC23A2Solute carrier family 23 member 2Transporter
SLC4A10Solute carrier family 4 member 10Transporter
SLCO6A1Solute carrier organic anion transporter family member 6A1Transporter
SND1Staphylococcal nuclease and tudor domain containing 1Transcription factor
SOAT1Sterol O-acyltransferase 1Enzyme
SOD1Superoxide dismutase 1Enzyme
SOD2Superoxide dismutase 2Enzyme
SPARCSecreted protein acidic and cysteine richSignaling
SPOCK1SPARC (osteonectin), cwcv and kazal like domains proteoglycan 1Enzyme modulator
SPP1Secreted phosphoprotein 1
SPRR2ASmall proline rich protein 2A
SPZ1Spermatogenic leucine zipper 1
SRBD1S1 RNA binding domain 1Nucleic acid binding
SRLSarcalumeninEnzyme modulator
SRSF3Serine and arginine rich splicing factor 3Nucleic acid binding
STIP1Stress induced phosphoprotein 1
SULT1E1Sulfotransferase family 1E member 1
TAP1Transporter 1, ATP binding cassette subfamily B memberTransporter
TBK1TANK binding kinase 1Kinase
TEKTEK receptor tyrosine kinaseKinase
TGFB1Transforming growth factor beta 1Signaling
TGFB2Transforming growth factor beta 2Signaling
TGFB3Transforming growth factor beta 3Signaling
TGFBR3Transforming growth factor beta receptor 3
TGM2Transglutaminase 2Enzyme
THBS1Thrombospondin 1
THBS2Thrombospondin 2
TIMP1TIMP metallopeptidase inhibitor 1Enzyme modulator
TIMP2TIMP metallopeptidase inhibitor 2Enzyme modulator
TIMP3TIMP metallopeptidase inhibitor 3Enzyme modulator
TIMP4TIMP metallopeptidase inhibitor 4Enzyme modulator
TLR2Toll like receptor 2
TLR4Toll like receptor 4
TMCO1Transmembrane and coiled-coil domains 1
TMTC2Transmembrane O-mannosyltransferase targeting cadherins 2
TNFTumor necrosis factorSignaling
TNNT1Troponin T1, slow skeletal typeCellular structure
TP53Tumor protein p53Transcription factor
TP53BP2Tumor protein p53 binding protein 2Enzyme modulator
TPX2TPX2 microtubule nucleation factorCellular structure
TRPM5Transient receptor potential cation channel subfamily M member 5Ion channel
TXNRD2Thioredoxin reductase 2Enzyme
URODUroporphyrinogen decarboxylase
USO1USO1 vesicle transport factorTransporter
VAV2Vav guanine nucleotide exchange factor 2
VAV3Vav guanine nucleotide exchange factor 3
VDRVitamin D receptorNuclear receptor
VEGFAVascular endothelial growth factor ASignaling
VEGFCVascular endothelial growth factor CSignaling
WDR36WD repeat domain 36
XRCC1X-ray repair cross complementing 1
ZNF410Zinc finger protein 410Transcription factor
ZNF469Zinc finger protein 469
ZP4Zona pellucida glycoprotein 4

Table 6.

Primary open angle glaucoma associated genes.

Literature reports that the potential therapeutic targets based on the molecular and cellular alterations caused by MYOC, OPTN and TBK1 mutations. Additionally, GWAS study performed in adult-onset glaucoma have identified novel loci for POAG (primary open-angle glaucoma) in CAV1/CAV2, CDKN2BAS, TMCO1, SIX6, 8q22(NTG), ABCA1, AFAP1, GMDS, PMM2, TGFBR3, FNDC3B, ARHGEF12, GAS7, FOXC1, ATXN2, TXNRD2, OPTC, MPP7 genes. Additionally, Single SNPs in the MYOC, COL8A2, COL1A1 and ZNF469 gene regions were reported by the study conducted in South Africa in POAG subjects.

GeneVariantChrPositionConsequenceAllelesClass
ABCB1rs74315329787509329Synonymous variantA/G;Tsnv
ABOrs289396889133262254Intron variantC/Tsnv
ADAMTS10rs75654767198589505Missense variantC/Tsnv
ADRB2rs10575193785148826910Stop gainedG/C;Tsnv
ADRB2rs13468658055148827322Missense variantC/Tsnv
AGERrs137854858632183666Missense variantC/Tsnv
APBB2rs137854860440995241Intron variantT/Csnv
APEX1rs1378548631420456995Missense variantT/A;C;Gsnv
ASB10rs1390067527151181233Synonymous variantG/Asnv
ASB10rs15535344217151181278Synonymous variantG/Asnv
ASB10rs15559542847151181173Synonymous variantC/A;G;Tsnv
ATOH7rs156435496810682319925 prime UTR variantT/Gsnv
ATOH7rs20014876410682320965 prime UTR variantA/Gsnv
B4GALT3;PPOXrs2007100761161175160Missense variantC/Tsnv
BIRC6rs201794655232545090Intron variantA/Tsnv
C14orf39;SIX6rs3734253951460509819Missense variantC/A;Gsnv
C14orf39;SIX6rs7506432161460509783Missense variantG/Asnv
C1orf112;SELErs8788544081169728058Missense variantC/A;Tsnv
CARD10rs12176910632237516037Missense variantC/A;Tsnv
CARD10rs21652412237508609Missense variantG/Asnv
CARD10rs339123452237508568Missense variantC/Tsnv
CARD10rs38259422237492794Missense variantG/Asnv
CARD10rs10486612237506365Missense variantG/Asnv
CATrs5662890991134438684Upstream gene variantC/Tsnv
CATrs10631921134461361Synonymous variantC/Tsnv
CATrs112581941134438925Upstream gene variantA/Tsnv
CAV1rs743153307116550415Intron variantG/Asnv
CAV2rs104252271165083163 prime UTR variantT/C;Gsnv
CD48rs129944011160681172Frameshift variantC/-;CC;CCCdelins
CD48rs1378532771160681173Frameshift variant-/Tins
CDKN2B;CDKN2B-AS1rs21493569220033683 prime UTR variantG/A;Tsnv
CDKN2B-AS1rs4986791922056500Intron variantG/Asnv
CDKN2B-AS1rs74315337922033367Non coding transcript exon variantC/Tsnv
CDKN2B-AS1rs10120688922062135Intron variantG/Tsnv
CDKN2B-AS1rs1131691014922055049Intron variantA/G;Tsnv
CDKN2B-AS1rs145285325922019130Intron variantA/Gsnv
CDKN2B-AS1rs2157719922028802Intron variantA/Gsnv
CNTNAP4rs22349261676307609Intron variantA/Gsnv
COL8A2rs25487136099217Missense variantC/G;Tsnv
COX1;COX2rs367923973MT6150Missense variantG/Asnv
COX2;COX1rs4898MT6253Missense variantT/Csnv
COX2;COX1;ATP8rs4986790MT6480Missense variantG/Asnv
CYP1B1rs74315328238071060Missense variantG/Csnv
CYP1B1rs74315332238071007Missense variantA/G;Tsnv
CYP1B1rs74315339238070996Missense variantT/C;Gsnv
CYP1B1;CYP1B1-AS1rs781662103238075034Missense variantC/Asnv
CYP1B1;CYP1B1-AS1rs878854066238075247Missense variantG/Csnv
CYP1B1;CYP1B1-AS1rs1001179238076389Non coding transcript exon variantG/Asnv
CYP1B1;CYP1B1-AS1rs10202118238075148Missense variantA/Tsnv
CYP1B1-AS1;CYP1B1rs1056827238074704Missense variantC/Tsnv
CYP46A1rs116566961499691630Non coding transcript exon variantA/Gsnv
DCLK1rs12796831336078480Intron variantT/Csnv
DDX3Xrs1533428X41346607Missense variantC/Tsnv
EDNRArs1694741475426883 prime UTR variantG/A;Csnv
ENO4rs1757610116864069Intron variantG/Asnv
ESR1rs17997506151929945Intron variantC/Asnv
ESR1rs17999836151970431Intron variantC/Asnv
ESR2rs1800401464279461Intron variantG/A;Tsnv
ESR2rs19000041464292158Intron variantC/Tsnv
FASLGrs1997528601172658358Upstream gene variantC/Tsnv
FASTKD1rs22349272169531449Missense variantA/Tsnv
FDXRrs2676069291774872110Stop gainedG/A;Csnv
FDXRrs32194891774863112Missense variantC/A;G;Tsnv
FNDC3Brs3694106163172315221Intron variantC/Gsnv
FNDC3Brs39181883172274597Intron variantG/Asnv
GAS7rs3975074441710130362Intron variantC/Asnv
GCM1rs74315334653258320Regulatory region variantT/Csnv
GPX4rs74315336191101993Upstream gene variantA/Gsnv
GSTP1rs743153381167585218Missense variantA/Gsnv
HSP90AA1rs75420314102083827Missense variantT/Csnv
IL20RArs7542373766137008718Missense variantG/Asnv
IL20RB-AS1;IL20RBrs100123136982255Missense variantC/Tsnv
INKA2;DDX20;LOC101928718rs100381771111754860Non coding transcript exon variantA/Tsnv
KCNQ4rs10063949140814886Intron variantC/Tsnv
KLC3;ERCC2rs10119701945351661Stop gainedT/A;Gsnv
LINC02640rs10427141068241124Intron variantC/Tsnv
LOC102724808;OPA1rs104519413193647160Missense variantA/Gsnv
LOC105376196rs10456429104933567Downstream gene variantG/Asnv
LOC107986513;GMDSrs10488647861707020Intron variantC/Tsnv
LOC112268121;EDNRB-AS1rs10519931377800045Intron variantA/Tsnv
LOC730100rs1052133251845108Intron variantC/Tsnv
LOC730100rs1052990251723186Intron variantC/Tsnv
LOC730100rs1056836251732120Intron variantT/A;Csnv
LOC730100rs1056837251725011Intron variantG/Asnv
LOXL1;LOXL1-AS1rs111253751573927241Missense variantG/A;C;Tsnv
LOXL1-AS1;LOXL1rs1116989341573929861Intron variantT/Csnv
LOXL1-AS1;LOXL1rs112410951573927205Missense variantG/Tsnv
LTBP2rs1129838581474551266Missense variantC/A;Tsnv
LTBP2rs11304091474502911Missense variantC/G;Tsnv
LTBP2rs11358401474505102Missense variantT/Csnv
MIR182;LOC105375501rs115368897129770387Non coding transcript exon variantC/G;Tsnv
MIR34C;BTG4;MIR34B;LOC728196rs1156865811111511840Intron variantT/Csnv
MMP9rs116699772046011586Missense variantA/Gsnv
MPP7rs11710635441028116482Intron variantG/C;Tsnv
MTHFRrs11720822111796309Missense variantA/Gsnv
MTHFRrs11771443111794407Missense variantT/Gsnv
MUL1rs12025126120503285Missense variantC/Tsnv
MUTYHrs12154178145331833Missense variantC/A;Gsnv
MUTYHrs121909194145329400Missense variantC/Tsnv
MYOCrs123776321171652476Stop gainedG/A;C;Tsnv
MYOCrs12554286051171652385Missense variantC/Tsnv
MYOCrs12560311171652468Missense variantC/Asnv
MYOCrs12686561171652578Missense variantC/Gsnv
MYOCrs12708417231171652341Stop gainedG/Asnv
MYOCrs127893791171652139Missense variantC/G;Tsnv
MYOCrs12793861171652539Missense variantA/Gsnv
MYOC;MYOCOSrs13155382741171636338Stop gainedG/Asnv
MYOC;MYOCOSrs131811171636382Missense variantG/Asnv
MYOC;MYOCOSrs131869121171636131Missense variantA/Gsnv
MYOC;MYOCOSrs140351171636143Missense variantA/Gsnv
MYOC;MYOCOSrs14287581171636161Missense variantC/Tsnv
MYOC;MYOCOSrs1434131161171636302Missense variantC/Gsnv
MYOC;MYOCOSrs1442498081171636185Missense variantT/Csnv
MYOC;MYOCOSrs1454372031171636010Missense variantA/C;Tsnv
MYOC;MYOCOSrs14634615891171636686Missense variantC/Tsnv
MYOC;MYOCOSrs14664415871171636542Missense variantC/Tsnv
MYOCOS;MYOCrs1467378471171636329Missense variantA/Gsnv
MYOCOS;MYOCrs1668501171636310Missense variantG/Asnv
MYOCOS;MYOCrs16951171636331Missense variantG/Asnv
MYOCOS;MYOCrs169842991171636000Missense variantG/Tsnv
MYOCOS;MYOCrs17997821171638703Missense variantG/A;Csnv
MYOCOS;MYOCrs18004401171636341Missense variantC/Tsnv
MYOCOS;MYOCrs18007791171636173Missense variantT/Csnv
MYOCOS;MYOCrs18008881171638675Missense variantC/Tsnv
MYOCOS;MYOCrs1845610871171636028Missense variantT/Csnv
MYOCOS;MYOCrs1858151461171635999Missense variantG/Asnv
NCKAP5rs18840542133605461Regulatory region variantT/A;C;Gsnv
ND2;RNR2;ND1rs1926320MT3010Non coding transcript exon variantG/Asnv
NDUFA6-DT;CYP2D6rs19279112242127941Missense variantG/A;Tsnv
NDUFA6-DT;CYP2D6rs1973882242126611Missense variantC/Gsnv
NOS3rs1994761287150999023Missense variantT/A;Gsnv
NOS3rs1997468247151005693Intron variantC/A;Tsnv
NOS3rs2001657367150990599Upstream gene variantC/Tsnv
NOS3rs2005476137150992855Intron variantG/A;Csnv
NOS3rs20706007150998107Intron variantG/Asnv
NOS3;ATG9Brs215632371510124833 prime UTR variantG/Tsnv
NRP1rs22535921033221802Missense variantA/Gsnv
NTF4rs23832041949060867Non coding transcript exon variantA/Gsnv
NTF4rs24724931949061660Missense variantA/Gsnv
NTF4rs25672061949061735Missense variantG/Asnv
NTF4rs27545111949061453Missense variantG/Asnv
NTMrs280121911131422069Intron variantA/G;Tsnv
OGG1rs2835858039756778Missense variantC/Tsnv
OGG1;CAMK1rs284298039757089Missense variantC/Gsnv
OPA1rs345512533193637313Intron variantT/A;Csnv
OPA1rs345952523193637285Splice region variantT/A;Csnv
OPTNrs37663551013109270Missense variantG/Asnv
OPTNrs37933421013136766Missense variantG/Asnv
OPTNrs38019941013110416Missense variantG/Csnv
OPTNrs3867410441013109279Frameshift variant-/AGCTdelins
OPTNrs39283061013109198Missense variantC/A;G;Tsnv
OPTNrs48801013132122Missense variantA/Gsnv
OPTNrs49387231013110400Missense variantT/Asnv
OPTNrs5230961013132098Missense variantA/Gsnv
OPTNrs5237471013110394Missense variantG/A;Tsnv
OPTNrs53351013124076Missense variantG/A;Csnv
OPTNrs5375168221013124076Missense variantG/A;Csnv
PBX2;AGERrs5479846321856573 prime UTR variantC/Asnv
PDIA5rs5542358973123150194Intron variantC/Tsnv
PRPF8rs571448378171684534Missense variantG/Asnv
PRPF8rs5743704171684498Missense variantA/Gsnv
PTGFRrs5746136178491756Intron variantC/A;Tsnv
RANrs576499843121308766963 prime UTR variantC/Tsnv
RERErs577370418699495Intron variantT/Csnv
RFTN1;OXNAD1rs57865060316354161Intron variantC/G;Tsnv
RHOArs59892895349363049Intron variantT/Csnv
RNR2;ND1rs61732310MT2416Non coding transcript exon variantT/Csnv
SEC14L2rs618547822230406040Intron variantC/A;G;Tsnv
SLC23A1rs64450555139383837Intron variantT/Csnv
SLC23A2rs690037205002446Intron variantG/A;Csnv
SNORD13G;ABCC4rs69175891395210754Missense variantC/Asnv
SOD2rs69342161596790843 prime UTR variantA/Tsnv
SOD2rs69940766159692840Missense variantA/Gsnv
SOD2rs703711761596820523 prime UTR variantC/Tsnv
SOD2rs704910561596782283 prime UTR variantT/Csnv
STIP1rs71594621164195658Missense variantC/A;Gsnv
STIP1rs7358601164203143Missense variantA/Gsnv
SYNE2;ESR2rs7377231464180928Intron variantT/Gsnv
TIMP1;SYN1;MIR4769rs74315331X47585586Synonymous variantT/Csnv
TLR2rs743153414153704799Missense variantC/Asnv
TLR4rs7464184069117711921Intron variantT/Gsnv
TLR4rs7467021109117713324Missense variantC/Tsnv
TLR4rs7470586339117713024Missense variantA/G;Tsnv
TLR4rs74778291177158533 prime UTR variantG/Csnv
TLR4rs74815149117710452Intron variantT/A;Csnv
TLR4rs7486214619117707776Intron variantA/Gsnv
TLR4rs74889994491177213853 prime UTR variantA/Gsnv
TMTC2rs7514179851282698057Intron variantG/Asnv
TP53rs751497460177676154Missense variantG/C;Tsnv
TP53rs754829637177676154Frameshift variant-/Cins
TP53rs755246983177676153Missense variantGG/ACmnv
TRPM5rs757228112415234Missense variantC/A;Tsnv
TTPArs75864656863087002Upstream gene variantA/Tsnv
TXNRD2rs758856722198760703 prime UTR variantT/Csnv
VAV2rs7618756129133855699Intron variantG/Asnv
VAV3rs7630682441107874935Missense variantG/Asnv
VAV3rs7631101107959790Intron variantC/Asnv
VAV3rs764817761107617607Missense variantA/C;Gsnv
WDR36rs7661471425111100751Intron variantC/Tsnv
WDR36rs7692175111103810Missense variantA/C;Gsnv
WDR36rs7820069655111121006Synonymous variantA/Tsnv
WDR36rs78305111092362Missense variantT/Csnv
WDR36rs79166975111119021Missense variantA/Gsnv
WTAPP1;MMP1rs791685211102797141Synonymous variantA/Gsnv
WTAPP1;MMP1rs794331611102799765Intron variantC/-delins
XRCC1rs79619531943551574Missense variantT/Csnv
XRCC1rs81766931943553422Missense variantG/Asnv
rs8790539141597027933Intergenic variantT/Csnv
rs92826711237933586Intergenic variantA/Csnv
rs95030121237935790Downstream gene variantT/A;Gsnv
rs9744951147919373Intergenic variantT/Csnv

Table 7.

Variants associated with primary open angle glaucoma.

GeneGene_Full_NameProtein_Class
ABCA1ATP binding cassette subfamily A member 1Transporter
ABCC5ATP binding cassette subfamily C member 5Transporter
ACDACD shelterin complex subunit and telomerase recruitment factor
AKR1C4Aldo-keto reductase family 1 member C4Enzyme
APOEApolipoprotein E
AQP1Aquaporin 1 (Colton blood group)Ion channel
ATOH7Atonal bHLH transcription factor 7Enzyme
BIRC6Baculoviral IAP repeat containing 6
BRCA1BRCA1 DNA repair associatedEnzyme
C10orf53Chromosome 10 open reading frame 53
C3Complement C3Enzyme modulator
CALCRLCalcitonin receptor like receptorG-protein coupled receptor
CATCatalaseEnzyme
CCL2C-C motif chemokine ligand 2Signaling
CCN2Cellular communication network factor 2Signaling
CDC42Cell division cycle 42Enzyme modulator
CDR1Cerebellar degeneration related protein 1
CHATCholine O-acetyltransferaseEnzyme
CIAO3Cytosolic iron-sulfur assembly component 3Enzyme
COL11A1Collagen type XI alpha 1 chain
COL1A1Collagen type I alpha 1 chain
CST3Cystatin C
CTSDCathepsin DEnzyme
CYP1B1Cytochrome P450 family 1 subfamily B member 1Enzyme
CYP2B6Cytochrome P450 family 2 subfamily B member 6
DBN1Drebrin 1Cellular structure
DCNDecorin
DPM2Dolichyl-phosphate mannosyltransferase subunit 2, regulatory
EIF2DEukaryotic translation initiation factor 2DReceptor
ELNElastin
EPDR1Ependymin related 1
F2Coagulation factor II, thrombinEnzyme
FAM102AFamily with sequence similarity 102 member A
FBLN7Fibulin 7
FERMT2Fermitin family member 2
GLIS3GLIS family zinc finger 3
GSTM1Glutathione S-transferase mu 1
HGFHepatocyte growth factorEnzyme
HLA-DPA1Major histocompatibility complex, class II, DP alpha 1Immune response
HSPA4Heat shock protein family A (Hsp70) member 4
HTR3C5-hydroxytryptamine receptor 3CIon channel
HTR3D5-hydroxytryptamine receptor 3DIon channel
IL1AInterleukin 1 alpha
IL1BInterleukin 1 beta
KDRKinase insert domain receptorKinase
KERAKeratocan
LGTNLigatin
LOC110599580CYP1B1 promoter
LOXLysyl oxidase
LOXL1Lysyl oxidase like 1
LOXL2Lysyl oxidase like 2
MFRPMembrane frizzled-related proteinEnzyme
MINDY4MINDY lysine 48 deubiquitinase 4
MMP1Matrix metallopeptidase 1Enzyme
MMP9Matrix metallopeptidase 9Enzyme
MTHFRMethylenetetrahydrofolate reductase
MYOCMyocilinCellular structure
NEBNebulin
NOS3Nitric oxide synthase 3
NT5E5'-nucleotidase ectoEnzyme
NTF4Neurotrophin 4Signaling
OGG18-oxoguanine DNA glycosylase
OPA3Outer mitochondrial membrane lipid metabolism regulator OPA3
OPTNOptineurin
PACC1Proton activated chloride channel 1
PARLPresenilin associated rhomboid likeEnzyme
PCMTD1Protein-L-isoaspartate (D-aspartate) O-methyltransferase domain containing 1Enzyme
PDE5APhosphodiesterase 5A
PDIA5Protein disulfide isomerase family A member 5
PLEKHA7Pleckstrin homology domain containing A7
PLXNA2Plexin A2
PRSS56Serine protease 56
RAC1Rac family small GTPase 1Enzyme modulator
RUNX1T1RUNX1 partner transcriptional co-repressor 1Transcription factor
SFRP4Secreted frizzled related protein 4
SOD2Superoxide dismutase 2Enzyme
SPARCSecreted protein acidic and cysteine richSignaling
SPP1Secreted phosphoprotein 1
ST18ST18 C2H2C-type zinc finger transcription factorTranscription factor
TGFB1Transforming growth factor beta 1Signaling
TGFB2Transforming growth factor beta 2Signaling
THBS1Thrombospondin 1
TMCO1Transmembrane and coiled-coil domains 1
TP53Tumor protein p53Transcription factor
TXNRD2Thioredoxin reductase 2Enzyme
VSX2Visual system homeobox 2
ZNRF3Zinc and ring finger 3

Table 8.

Genes associated with primary angle closure glaucoma.

The inheritance pattern of angle closure causing mutations in COL18A1 was autosomal dominant. PACG (primary angle-closure glaucoma (EPDR1, CHAT, GLIS3, FERMT2, DPM2-FAM102); and exfoliation syndrome (XFS) glaucoma (CACNA1A). Additionally, it has been reported that the most significant GWAS in the Asian population were identified in SNPs of rs11024102 (PLEKHA7; 11p15.1), rs3753841 (COL11A1; 1p21.1), and rs1015213 (8q11.23).

GeneVariantChrPositionConsequenceAllelesClass
ABCC5rs11327763183978614Synonymous variantA/Gsnv
ABCC5rs9393363183967746Stop gainedA/G;Tsnv
BIRC6rs2754511232545090Intron variantA/Tsnv
C10orf53rs12582671049687724Intron variantG/Asnv
CALCRL;LOC105373786rs11576992187394177Intron variantC/G;Tsnv
CATrs10011791134438684Upstream gene variantC/Tsnv
COL11A1rs37538411102914362Missense variantG/Asnv
COL11A1rs121389771102927901Intron variantC/Tsnv
COL11A1rs16764861102888582Missense variantA/G;Tsnv
DPM2;FAM102Ars37398219127940198Non coding transcript exon variantA/Gsnv
EPDR1;SFRP4rs3816415737948709Intron variantG/Asnv
FERMT2rs74943791452944673Intron variantC/G;Tsnv
GLIS3rs73689394217028Intron variantG/A;Csnv
HGFrs17427817781735119Intron variantC/A;G;Tsnv
HGFrs12540393781734871Intron variantC/Tsnv
HGFrs3735520781771623Upstream gene variantG/A;Tsnv
HGFrs5745718781718232Intron variantT/Gsnv
HSPA1L;HSPA1Ars10436186318157305 prime UTR variantG/A;C;Tsnv
HTR3Drs124935503184034985Intron variantG/Asnv
LINC02640rs19000041068241124Intron variantC/Tsnv
LOC105373786;CALCRLrs67595352187373374Intron variantT/Csnv
LOC105373786;CALCRLrs8406172187365606Intron variantA/Tsnv
LOC107985096rs16764841102839465Intron variantA/Csnv
LOXL1;LOXL1-AS1rs38259421573927241Missense variantG/A;C;Tsnv
LOXL1-AS1;LOXL1rs21652411573929861Intron variantT/Csnv
MMP1;WTAPP1rs75645909411102795237Missense variantT/A;C;Gsnv
MMP9rs175762046011586Missense variantA/Gsnv
MMP9rs26645382046011586Missense variantA/Gsnv
MMP9rs39182492046009497Intron variantT/Csnv
MTHFRrs1217691063111796309Missense variantA/Gsnv
MYOCrs1835321171640341Intron variantT/A;Csnv
MYOCrs2358751171644616Intron variantC/Tsnv
MYOCrs2359131171649516Intron variantT/C;Gsnv
NOS3rs37933427150998107Intron variantG/Asnv
NTF4rs116699771949060867Non coding transcript exon variantA/Gsnv
NTF4rs617323101949061735Missense variantG/Asnv
PDIA5rs117208223123150194Intron variantC/Tsnv
PLEKHA7rs110241021116987058Intron variantT/Csnv
PLEKHA7rs2164891116802189Intron variantG/A;Tsnv
SLC12A5-AS1;MMP9rs22508892046013767Missense variantG/C;Tsnv
SLC12A5-AS1;MMP9rs175772046014472Missense variantG/A;Csnv
SLC38A4rs9836671246769523Intron variantC/Tsnv
SOD2rs48806159692840Missense variantA/Gsnv
TP53rs1042522177676154Missense variantG/C;Tsnv
TP53rs1131691014177676154Frameshift variant-/Cins
TP53rs878854066177676153Missense variantGG/ACmnv
TXNRD2rs37883172219902302Intron variantG/Tsnv
VAV2rs21563239133855699Intron variantG/Asnv
VAV3rs14664415871107874935Missense variantG/Asnv
VAV3rs28012191107959790Intron variantC/Asnv
VAV3rs5764998431107617607Missense variantA/C;Gsnv
WTAPP1;MMP1rs179975011102799765Intron variantC/-delins
ZNRF3rs729011722290548683 prime UTR variantC/G;Tsnv
rs1015213851974981Intron variantC/Tsnv
rs46564611165717968TF binding site variantG/A

Table 9.

Variants associated with primary angle closure glaucoma.

Advertisement

8. Conclusions

Glaucoma genetics and genomics have to be assessed with the larger picture of visual impairment, disease prevalence, comorbidities, genetics, genomics, disease mechanisms, mechanical stress, neuroprotection, neurodegeneration, apoptosis, and immune imbalance. Few single causative genes, but multiple genes’ dysregulated expressions at several tissues’ sites of the eye like ciliary body, trabecular meshwork, lamina cribrosa, retina and optic nerve determine the spectrum of phenomics in glaucoma (Figure 1). This has led to the identification of neurotrophic factors, and anti-apoptotic molecules to prevent further neurodegeneration of RGCs and loss of vision. The complex nature of the disease and the discovery of several hundred genes and molecules is a boon and a bane at the same time. This status needs further research to focus and identify a battery of few molecules that could be used, individually or as a cocktail, in a majority of patients with glaucoma. However, it looks like the field may move towards a cocktail of molecular therapy based on personalised medicine and the individuals’ genetic signature pattern and phenomics.

Figure 1.

The ocular tissues, genomics and biomechanisms of glaucoma.

References

  1. 1. Cueto AFV, Álvarez L, García M, Álvarez-barrios A, Artime E, Cueto LFV, et al. Candidate glaucoma biomarkers: From proteins to metabolites, and the pitfalls to clinical applications. Biology. 2021;10:763
  2. 2. Zukerman R, Harris A, Oddone F, Siesky B, Verticchio Vercellin A, Ciulla TA. Glaucoma heritability: Molecular mechanisms of disease. Genes (Basel). 2021;12:8
  3. 3. Roughead EE, Kalisch LM, Pratt NL, Killer G, Barnard A, Gilbert AL. Managing glaucoma in those with co-morbidity: Not as easy as it seems. Biology. 2021;19:74
  4. 4. Flaxman SR, Bourne RRA, Resnikoff S, Ackland P, Braithwaite T, Cicinelli M V, et al. Articles Global causes of blindness and distance vision impairment 1990-2020: A systematic review and meta-analysis. 2017. Available from: www.thelancet.com/lancetgh
  5. 5. (17) (PDF) World blindness and visual impairment: Despite many successes, the problem is growing [Internet]. [cited 2020 Apr 6]. Available from: https://www.researchgate.net/publication/323435449_World_blindness_and_visual_impairment_despite_many_successes_the_problem_is_growing
  6. 6. Gordois A, Cutler H, Pezzullo L, Gordon K, Cruess A, Winyard S, et al. An estimation of the worldwide economic and health burden of visual impairment. Global Public Health. 2012;7(5):465-481
  7. 7. Wittenborn JS, Zhang X, Feagan CW, et al. The economic burden of vision loss and eye disorders among the United States population younger than 40 years. Ophthalmology. 2013;120(9):1728-1735
  8. 8. Bourne RR, Stevens GA, White RA, et al. Causes of vision loss worldwide, 1990-2010: A systematic analysis. Lancet Global Health. 2013;1(6):e339-e349
  9. 9. Khanna RC, Marmamula S, Rao GN. International vision care: Issues and approaches. Annual Review of Visual Science. 2017;3(1):53-68
  10. 10. Raman R, Rani PK, Reddi Rachepalle S, et al. Prevalence of diabetic retinopathy in India: Sankara Nethralaya diabetic retinopathy epidemiology and molecular genetics study report 2. Ophthalmology. 2009;116(2):311-318
  11. 11. Zetterberg M. Age-Related Eye Disease and Gender. Maturitas: Elsevier Ireland Ltd; 2016. pp. 19-26
  12. 12. Stevens GA, White RA, Flaxman SR, Price H, Jonas JB, Keeffe J, et al. Global prevalence of vision impairment and blindness: Magnitude and temporal trends, 1990-2010. Ophthalmology. 2013;120(12):2377-2384
  13. 13. Reitmeir P, Linkohr B, Heier M, Molnos S, Strobl R, Schulz H, et al. Common eye diseases in older adults of southern Germany: results from the KORA-Age study. National Health Aging Trends in Study Pain. 2011;154(12):481-486
  14. 14. Wong TY, Loon SC, Saw SM. The epidemiology of age related eye diseases in Asia [Internet]. British Journal of Ophthalmology. 2006;90:506-511
  15. 15. Hashemi H, Khabazkhoob M, Nabovati P, Ostadimoghaddam H, Shafaee S, Doostdar A, et al. The prevalence of age-related eye disease in an elderly population. Ophthalmic Epidemiology. 2017;24(4):222-228
  16. 16. Klein R, Klein BEK. The prevalence of age-related eye diseases and visual impairment in aging: Current estimates. Investigative Ophthalmology and Visual Science. 2013;54(14):SF5
  17. 17. Maberley DA, Hollands H, Chuo J, et al. The prevalence of low vision and blindness in Canada. Eye (Lond). 2006;20(3):341-346. DOI: 10.1038/sj.eye.6701879
  18. 18. Congdon N. Causes and prevalence of visual impairment among adults in the United States. Archives of Ophthalmology. 2004;122(4):477-485
  19. 19. Chou R, Dana T, Bougatsos C, Grusing S, Blazina I. Screening for impaired visual acuity in older adults: Updated evidence report and systematic review for the US preventive services task force. JAMA. 2016;315(9):915-933
  20. 20. Prasad S, Kamath GG, Jones K, Clearkin LG, Phillips RP. Prevalence of blindness and visual impairment in a population of people with diabetes. Eye. 2001;15(5):640-643
  21. 21. Sheeladevi S, Seelam B, Nukella P, Borah R, Ali R, Keay L. Prevalence of refractive errors, uncorrected refractive error, and presbyopia in adults in India: A systematic review. Indian Journal of Ophthalmology. 2019;67:583-592
  22. 22. Furtado JM, Lansingh VC, Carter MJ, Milanese MF, Peña BN, Ghersi HA, et al. Causes of blindness and visual impairment in Latin America. Survey of Ophthalmology. 2012;57(2):149-177
  23. 23. Zheng Y, Lavanya R, Wu R, Wong WL, Wang JJ, Mitchell P, et al. Prevalence and causes of visual impairment and blindness in an urban Indian population: The Singapore Indian eye study. Ophthalmology. 2011;118(9):1798-1804
  24. 24. Liang YB, Friedman DS, Wong TY, et al. Prevalence and causes of low vision and blindness in a rural Chinese adult population: The Handan Eye Study. Ophthalmology. 2008;115(11):1965-1972
  25. 25. Chiang PPC, Zheng Y, Wong TY, Lamoureux EL. Vision impairment and major causes of vision loss impacts on vision-specific functioning independent of socioeconomic factors. Ophthalmology. 2013;120(2):415-422
  26. 26. Ramke J, Zwi AB, Palagyi A, Blignault I, Gilbert CE. Equity and blindness: Closing evidence gaps to support universal eye health. Ophthalmic Epidemiology. 2015;22(5):297-307
  27. 27. Hashemi N, Moghaddasi H, Rabiei R, Asadi F, Farahi A. Eye health information systems in selected countries. Journal of Ophthalmic Visual Research. 2018;13(3):333-338
  28. 28. Boerma T, Eozenou P, Evans D, Evans T, Kieny MP, Wagstaff A. Monitoring progress towards universal health coverage at country and global levels. PLoS Medicine. 2014;11(9):e1001731
  29. 29. Yang X, Pan X, Zhao X, Luo J, Xu M, Bai D, et al. Autophagy and Age-Related Eye Diseases. 2019
  30. 30. Paul S, Abraham V. How healthy is our geriatric population? A community-based cross-sectional study. Journal of Family Medicine and Primary Care. 2015;4(2):221
  31. 31. Vijaya L, George R, Arvind H, Baskaran M, Raju P, Ramesh SV, et al. Prevalence and causes of blindness in the rural population of the Chennai Glaucoma Study. British Journal of Ophthalmology. 2006;90(4):407
  32. 32. Quigley H, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. The British Journal of Ophthalmology. 2006;90:262-267
  33. 33. Tham Y-C, Li X, Wong TY, Quigley HA, Aung T, Cheng C-Y. Global Prevalence of Glaucoma and Projections of Glaucoma Burden through 2040 A Systematic Review and Meta-Analysis. Ophthalmology. 2014;121:2081
  34. 34. Bourne RRA. Worldwide glaucoma through the looking glass. The British Journal of Ophthalmology. 2020;90:253-254
  35. 35. Kapetanakis VV, MPY C, Foster PJ, Cook DG, Owen CG, Rudnicka AR. Global variations and time trends in the prevalence of primary open angle glaucoma (POAG): A systematic review and meta-analysis. The British Journal of Ophthalmology. 2016;100:86-93
  36. 36. Chiu SL, Chu CL, Muo CH, Chen CL, Lan SJ. The prevalence and the incidence of diagnosed open-angle glaucoma and diagnosed angle-closure glaucoma: Changes from 2001 to 2010. Journal of Glaucoma. 2016;25(5):e514-e519
  37. 37. George R, Ve RS, Vijaya L. Glaucoma in India: Estimated burden of disease. Journal of Glaucoma. 2010;19(6):391-397
  38. 38. Rein DB, Zhang P, Wirth KE, Lee PP, Hoerger TJ, McCall N, et al. The economic burden of major adult visual disorders in the United States. Archives of Ophthalmology. 2006;124(12):1754-1760
  39. 39. Leffler CT, Schwartz SG, Giliberti FM, Young MT, Bermudez D. What was Glaucoma Called before the 20th Century?: 2015
  40. 40. Teikari JM. Genetic factors in open-angle (simple and capsular) glaucoma: A population-based twin study. Acta Ophthalmologica. 1987;65(6):715-720
  41. 41. Cuellar-Partida G, Craig JE, Burdon KP, Wang JJ, Vote BJ, Souzeau E, et al. Assessment of polygenic effects links primary open-angle glaucoma and age-related macular degeneration. Scientific Reports. 2016;31:6
  42. 42. Ge T, Chen C-Y, Neale BM, Sabuncu MR, Smoller JW. Phenome-wide heritability analysis of the UK Biobank. PLOS Genetics. 2017;13(4):e1006711
  43. 43. Weinreb RN, Leung CK, Crowston JG, et al. Primary open-angle glaucoma. Nature Reviews Disease Primers. 2016;2:16067
  44. 44. Asefa NG, Neustaeter A, Jansonius NM, Snieder H. Heritability of glaucoma and glaucoma-related endophenotypes: Systematic review and meta-analysis. Survive in Ophthalmology. 2019;64(6):835-851
  45. 45. Cascella R, Strafella C, Germani C, et al. The genetics and the genomics of primary congenital Glaucoma. BioMed Research International. 2015;2015:321291
  46. 46. Sripriya S, George R, Vijaya L, Kumaramanickavel G. Basic science: Understanding the biology of Glaucoma: The current scenario. Current Journal of Glaucoma. 2007;1(1):7-16
  47. 47. Plášilová M, Stoilov I, Sarfarazi M, Kádasi L, Feráková E, Ferák V. Identification of a single ancestral CYP1B1 mutation in Slovak Gypsies (Roms) affected with primary congenital glaucoma. Journal of Medical Genetics. 1999;36(4):290-294
  48. 48. Dandona L, Williams JD, Williams BCRG. Population-based assessment of childhood blindness in southern India. Archives of Ophthalmology. 1998;116:545-546
  49. 49. Ramprasad VL, George RJ, Sripriya S, Nirmaladevi J, Vijaya L, Kumaramanickavel G. Molecular genetic analysis of a consanguineous south Indian family with congenital glaucoma: Relevance of genetic testing and counseling. Ophthalmic Genetics. 2007;28(1):17-24
  50. 50. Sripriya S, Nirmaladevi J, George R, et al. OPTN gene: Profile of patients with glaucoma from India. Molecular Vision. 2006;12:816-820
  51. 51. Stone EM, Fingert JH, Alward WLM, Nguyen TD, Polansky JR, Sunden SLF, et al. Identification of a gene that causes primary open angle glaucoma. Science. 1997;275(5300):668-670
  52. 52. Sripriya S, Uthra S, Sangeetha R, George RJ, Hemmamalini A, Paul PG, et al. Low frequency of myocilin mutations in Indian primary open-angle glaucoma patients. Clinical Genetics. 2004;65(4):333-337
  53. 53. Quigley HA, Stone EM, Fingert JH. Familial glaucoma—A pedigree revisited with genetic testing after 70 years. JAMA Ophthalmology. 2022;140(5):543-544
  54. 54. Cunha DL, Arno G, Corton M, Moosajee M. The spectrum of PAX6 mutations and genotype-phenotype correlations in the eye. Genes. 2019;10(12):1050
  55. 55. Wallace DJ, Chau FY, Santiago-Turla C, Hauser M, Challa P, Lee PP, et al. Osteogenesis imperfecta and primary open angle glaucoma: Genotypic analysis of a new phenotypic association. Molecular Visual. 2014;20:1174
  56. 56. Balikov DA, Jacobson A, Prasov L. Glaucoma syndromes: Insights into glaucoma genetics and pathogenesis from monogenic syndromic disorders. Genes. 2021;12:9
  57. 57. Souma T, Tompson SW, Thomson BR, Siggs OM, Kizhatil K, Yamaguchi S, et al. Angiopoietin receptor TEK mutations underlie primary congenital glaucoma with variable expressivity. Journal of Clinical Investment. 2016;126(7):2575
  58. 58. Berry FB, Lines MA, Oas JM, Footz T, Underhill DA, Gage PJ, et al. Functional interactions between FOXC1 and PITX2 underlie the sensitivity to FOXC1 gene dose in Axenfeld–Rieger syndrome and anterior segment dysgenesis. Human Molecular Genetics. 2006;15(6):905-919
  59. 59. Graw J. The genetic and molecular basis of congenital eye defects. Nature Reviews. Genetics. 2003;4(11):876-888
  60. 60. Ragge NK, Lorenz B, Schneider A, Bushby K, De Sanctis L, De Sanctis U, et al. SOX2 anophthalmia syndrome. American Journal of Medical Genetetics. 2005;135:1-7
  61. 61. Davidson AE, Cheong SS, Hysi PG, Venturini C, Plagnol V, Ruddle JB, et al. Association of CHRDL1 mutations and variants with X-linked Megalocornea, Neuhäuser syndrome and central corneal thickness. PLoS One. 2014;9:2012-2016
  62. 62. Shetty R, Nuijts RM, Nanaiah SG, et al. Two novel missense substitutions in the VSX1 gene: Clinical and genetic analysis of families with Keratoconus from India. BMC Medical Genetics. 2015;16:33
  63. 63. Reis LM, Semina EV. Genetics of anterior segment dysgenesis disorders. Current Opinion in Ophthalmology. 2011;22(5):314-324
  64. 64. DeJesus Y, Moreno Ceballos G. A timeline of discovery and current research on primary open-angle glaucoma and emergence of potentially permanent treatment solutions. Spectra Undergraduate Research Journal. 2021;1(2):24-35
  65. 65. Sudlow C, Gallacher J, Allen N, et al. UK Biobank: An open access resource for identifying the causes of a wide range of complex Diseases of middle and old Age. PLoS Medicine. 2015;12(3):e1001779
  66. 66. Choquet H, Paylakhi S, Kneeland SC, Thai KK, Hoffmann TJ, Yin J, et al. A multiethnic genome-wide association study of primary open-angle glaucoma identifies novel risk loci. Nature Communications. 2018;9(1):1-14
  67. 67. Burdon KP. Genome-wide association studies in the hunt for genes causing primary open-angle glaucoma: a review. Clinical Experiment Ophthalmology. 2012;40(4):358-363
  68. 68. Abu-Amero K, Kondkar A, Chalam K. An updated review on the genetics of primary open angle glaucoma. International Journal of Molecular Science. 2015;16(2):28886-28911
  69. 69. Miller MA, Fingert JH, Bettis DI. Genetics and genetic testing for glaucoma. Current Opinion in Ophthalmology. 2017;28:133-138
  70. 70. Liu Y, Allingham RR. Molecular genetics in glaucoma. Experimental Eye Research. Oct 2011;93(4):331-339
  71. 71. Wiggs JL, Hauser MA, Abdrabou W, Allingham RR, Budenz DL, Delbono E, et al. The NEIGHBOR consortium primary open angle Glaucoma genome-wide association study: Rationale, study design and clinical variables. Journal of Glaucoma. 2013;22(7):517
  72. 72. Verma SS, Cooke Bailey JN, Lucas A, Bradford Y, Linneman JG, Hauser MA, et al. Epistatic gene-based interaction analyses for Glaucoma in eMERGE and NEIGHBOR consortium. PLoS Genetics. 2016;12(9)
  73. 73. Sripriya S, George R, Arvind H, et al. Transforming growth factor beta-1 -509C>T polymorphism in Indian patients with primary open angle glaucoma. Molecular Diagnosis Therapy. 2007;11(3):151-154
  74. 74. Nair KS, Srivastava C, Brown RV, Koli S, Choquet H, Kang HS, et al. GLIS1 regulates trabecular meshwork function and intraocular pressure and is associated with glaucoma in humans. Nature Communication. 2021;12(1):15
  75. 75. Quigley HA, Congdon NG, Friedman DS. Glaucoma in China (and worldwide): Changes in established thinking will decrease preventable blindness. The British Journal of Ophthalmology. 2001;85:1271-1272
  76. 76. Vithana EN, Khor CC, Qiao C, Nongpiur ME, George R, Chen LJ, et al. Genome-wide association analyses identify three new susceptibility loci for primary angle closure glaucoma. Nature Genetics. 2012;44(10):1142-1146
  77. 77. Ahram DF, Alward WL, Kuehn MH. The genetic mechanisms of primary angle closure glaucoma. Eye. 2015;29(10):1251
  78. 78. Wiggs JL, Pasquale LR. Genetics of glaucoma. Human Molecular Genetics. 2017;26(R1):R21
  79. 79. Inamori Y, Ota M, Inoko H, Okada E, Nishizaki R, Shiota T, et al. The COL1A1 gene and high myopia susceptibility in Japanese. Human Genetics. 2007;122(2):151-157
  80. 80. Zhang D, Shi Y, Gong B, He F, Lu F, Lin H, et al. An association study of the COL1A1 gene and high myopia in a Han Chinese population. 2022
  81. 81. Aboobakar IF, Wiggs JL. The genetics of glaucoma: Disease associations, personalised risk assessment and therapeutic opportunities-A review. Clinical Experiment in Ophthalmology. 2022;50(2):143-162
  82. 82. Thorleifsson G, Magnusson KP, Sulem P, Walters GB, Gudbjartsson DF, Stefansson H, et al. Common sequence variants in the LOXL1 gene confer susceptibility to exfoliation glaucoma. Science. 2007;317(5843):1397-1400
  83. 83. Ramprasad VL, George R, Soumittra N, Sharmila F, Vijaya L, Kumaramanickavel G. Association of non-synonymous single nucleotide polymorphisms in the LOXL1 gene with pseudoexfoliation syndrome in India. Molecular Vision. 9 Feb 2008;14:318-322
  84. 84. Aboobakar IF, Johnson WM, Stamer WD, Hauser MA, Allingham RR. Major review: Exfoliation syndrome; advances in disease genetics, molecular biology, and epidemiology. Experimental Eye Research. 2017;154:88-103
  85. 85. Tian Y, Li J, Su S, Cao Y, Wang Z, Zhao S, et al. PCOS-GWAS susceptibility variants in THADA, INSR, TOX3, and DENND1A are associated with metabolic syndrome or insulin resistance in women with PCOS. Frontier in Endocrinology. 2020;11:274
  86. 86. Weinreb RN, Aung T, Medeiros FA. The pathophysiology and treatment of glaucoma: A review. JAMA. 2014;311(18):1901-1911
  87. 87. Dias MS, Araujo VG de, Lani-Louzada R, Linden R, Ribas VT, Petrs-Silva H. Perspective on Gene Therapy for Glaucoma. 2022
  88. 88. Prada D, Harris A, Guidoboni G, Siesky B, Huang AM, Arciero J. Autoregulation and neurovascular coupling in the optic nerve head. Survive in Ophthalmology. 2016;61(2):164-186
  89. 89. Haro E, Petit F, Pira CU, Spady CD, Lucas-Toca S, Yorozuya LI, et al. Identification of limb-specific Lmx1b auto-regulatory modules with Nail-patella syndrome pathogenicity. Natural Communication. 2021;12(1):1-11
  90. 90. Zukerman R, Harris A, Oddone F, Siesky B, Verticchio Vercellin A, Ciulla TA. Glaucoma heritability: Molecular mechanisms of disease. Genes. 2021;12(8):1135
  91. 91. Springelkamp H, Höhn R, Mishra A, Hysi PG, Khor CC, Loomis SJ, et al. Meta-analysis of genome-wide association studies identifies novel loci that influence cupping and the glaucomatous process. Nature Communication. 2014;5:47
  92. 92. Mamatha G, Srilekha S, Meenakshi S, Kumaramanickavel G. Screening of the RPE65 gene in the Asian Indian patients with leber congenital amaurosis. Ophthalmic Genetics. 2008;29(2):73-78
  93. 93. Faro V, Bhattacharya A, Zhou W, Zhou D, Wang Y, Läll K, et al. Genome-wide association meta-analysis identifies novel ancestry-specific primary open-angle glaucoma loci and shared biology with vascular mechanisms and cell proliferation. medRxiv. 2021;2021:10
  94. 94. Nickells RW. The cell and molecular biology of Glaucoma: Mechanisms of retinal ganglion cell death. Investigative Ophthalmology & Visual Science. 2012;53(5):2476-2481
  95. 95. Quigley HA, Nickells RW, Kerrigan LA, Pease ME, Thibault DJ, Zack DJ. Retinal ganglion cell death in experimental glaucoma and after axotomy occurs by apoptosis. Investigative Ophthalmology & Visual Science. 1995;36(5):774-786
  96. 96. Cordeiro MF, Normando EM, Cardoso MJ, Miodragovic S, Jeylani S, Davis BM, et al. Real-time imaging of single neuronal cell apoptosis in patients with glaucoma. Brain. 2017;140(6):1757-1767
  97. 97. Mead B, Kerr A, Nakaya N, Tomarev SI. miRNA changes in retinal ganglion cells after optic nerve crush and glaucomatous damage. Cells. 2021;10(7):1757
  98. 98. Pernet V, Di Polo A. BDNF promotes robust survival of retinal ganglion cells but not axon regeneration within the Adult rat optic nerve. Investigative Ophthalmology & Visual Science. 2003;44(13):136
  99. 99. R Z, A H, AV V, B S, LR P, TA C. Molecular genetics of glaucoma: Subtype and ethnicity considerations. Genes. 2020;12(1):1-36
  100. 100. Liu J, Gao HY, Wang XF. The role of the Rho/ROCK signaling pathway in inhibiting axonal regeneration in the central nervous system. Neural Regeneration Research. 2015;10(11):1892
  101. 101. Barros Ribeiro Da Silva V, Porcionatto M, Toledo Ribas V. The rise of molecules able to regenerate the central nervous system. Journal of Medical Chemistry. 2022;63(2):490-511
  102. 102. Reinehr S, Guntermann A, Theile J, Benning L, Grotegut P, Kuehn S, et al. Proteomic analysis of retinal tissue in an S100B autoimmune glaucoma model. Biology. 2021;11(1):16
  103. 103. Fudalej E, Justyniarska M, Kasarełło K, Dziedziak J, Szaflik JP, Cudnoch-Jȩdrzejewska A. Neuroprotective factors of the retina and their role in promoting survival of retinal ganglion cells: A review. Ophthalmic Research. 2021;64(3):345-355
  104. 104. Clark AF. The cell and molecular biology of Glaucoma: Biomechanical factors in Glaucoma. Investigative Ophthalmology & Visual Science. 2012;53(5):2473-2475
  105. 105. Piñero J, Saüch J, Sanz F, Furlong LI. The DisGeNET cytoscape app: Exploring and visualizing disease genomics data. Computational and Structural Biotechnology Journal. 2021;19:2960-2967

Written By

Govindasamy Kumaramanickavel, Siddhita Jadhav, Sugirdhana Parthiban Ramsait and Pinakin Gunvant Davey

Submitted: 09 August 2022 Reviewed: 21 October 2022 Published: 09 December 2022