glaucoma – diabetes of the brain: a radical hypothesis about its nature and pathogenesis

12
Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis Muneeb A. Faiq a,b,c , Rima Dada b , Daman Saluja c , Tanuj Dada a,a Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi 110029, India b Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, India c Medical Biotechnology Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi 110007, India article info Article history: Received 27 December 2013 Accepted 3 February 2014 abstract Glaucoma is the leading cause of irreversible blindness characterized by irremediable loss of retinal ganglion cells. Its risk increases with progressing age and elevated intraocular pressure. Studies have established that glaucoma is a neurodegenerative disorder in which the damage involves many brain tissues from retina to the lateral geniculate nucleus. Despite lot of research, complete pathomechanism of glaucoma is not known and there is no treatment available except modification of intraocular pressure pharmacologically and/or surgically. We here present a hypothesis inspired by studies across many areas of molecular and clinical sciences in an integrative manner that leads to a uniquely unconventional understanding of this disorder. Our hypothesis postulates that glaucoma may possibly be the diabetes of the brain. Based on the remarkable similarities between glaucoma and diabetes we propose glaucoma also to be a type of diabetes. Glaucoma and diabetes share many aspects from various molecular mech- anisms to involvement of insulin and possible use of antidiabetics in glaucoma therapy. Additionally, Alzheimer’s disease has already been proposed to be diabetes type-3. We show that Alzheimer’s disease is cerebral glaucoma and diabetes at the same time which, by transitive property of similarities, again leads to our hypothesis that glaucoma is diabetes of the brain. Our proposition may lead to appreciation of certain important facets of glaucoma which have previously not been given due consideration. It also may lead to an alternative classification of diabetes as pancreatic and brain diabetes thereby widening the vision arena of the understanding of both these disorders. Ó 2014 Elsevier Ltd. All rights reserved. If at first the idea is not absurd then there is no hope for it. Albert Einstein Hypothesis We hypothesize that ‘‘Glaucoma is the Diabetes of the Brain’’. Our proposition derives support from our arguments that (i) there are striking similarities in molecular biology of glaucoma and dia- betes, (ii) brain can also suffer from diabetes, (iii) glaucoma inflicts brain and the brain part of eye, (iv) insulin is involved in mainte- nance of brain function, intraocular pressure and presumably other ocular functions and (v) brain diabetes is caused by same reagents which cause pancreatic diabetes. This hypothesis is important be- cause it may lead to understanding of certain areas of glaucoma and diabetes which have not been evaluated yet. Our hypothesis is likely to give birth to new areas in glaucoma and diabetes under- standing and management. Glaucoma–diabetes: a molecular crosstalk Glaucoma is a progressive ocular neurodegeneration and sec- ond leading cause of blindness with more than 60 million people affected worldwide [1]. The hallmark of glaucoma is gradual loss of retinal ganglion cells (RGCs) [2] by apoptosis [3]. Glaucoma has certain molecular and mechanistic similarities with other degenerative disorders of the central nervous system (CNS) like Alzheimer’s disease (AD) and Parkinson’s disease (PD). Several pathways for RGC death have been observed in glaucoma [2] and insulin plays a key role in many of them. Factors implicated in glaucoma pathogenesis include rise in intraocular pressure (IOP), decreased production of neurotrophic factors, excitotoxicity and vasospastic hypoxia [4,5]. RGCs die in glaucoma by various combinations and permuta- tions of apoptosis, oxidative stress (OS), mitochondrial dysfunction, http://dx.doi.org/10.1016/j.mehy.2014.02.005 0306-9877/Ó 2014 Elsevier Ltd. All rights reserved. Corresponding author. Tel.: +91 11 26588500x3001; fax: +91 11 26588663. E-mail address: [email protected] (T. Dada). Medical Hypotheses 82 (2014) 535–546 Contents lists available at ScienceDirect Medical Hypotheses journal homepage: www.elsevier.com/locate/mehy

Upload: tanuj

Post on 30-Dec-2016

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

Medical Hypotheses 82 (2014) 535–546

Contents lists available at ScienceDirect

Medical Hypotheses

journal homepage: www.elsevier .com/locate /mehy

Glaucoma – Diabetes of the brain: A radical hypothesis about its natureand pathogenesis

http://dx.doi.org/10.1016/j.mehy.2014.02.0050306-9877/� 2014 Elsevier Ltd. All rights reserved.

⇑ Corresponding author. Tel.: +91 11 26588500x3001; fax: +91 11 26588663.E-mail address: [email protected] (T. Dada).

Muneeb A. Faiq a,b,c, Rima Dada b, Daman Saluja c, Tanuj Dada a,⇑a Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, Delhi 110029, Indiab Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi 110029, Indiac Medical Biotechnology Laboratory, Dr. B.R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi 110007, India

a r t i c l e i n f o a b s t r a c t

Article history:Received 27 December 2013Accepted 3 February 2014

Glaucoma is the leading cause of irreversible blindness characterized by irremediable loss of retinalganglion cells. Its risk increases with progressing age and elevated intraocular pressure. Studies haveestablished that glaucoma is a neurodegenerative disorder in which the damage involves many braintissues from retina to the lateral geniculate nucleus. Despite lot of research, complete pathomechanismof glaucoma is not known and there is no treatment available except modification of intraocular pressurepharmacologically and/or surgically. We here present a hypothesis inspired by studies across many areasof molecular and clinical sciences in an integrative manner that leads to a uniquely unconventionalunderstanding of this disorder. Our hypothesis postulates that glaucoma may possibly be the diabetesof the brain. Based on the remarkable similarities between glaucoma and diabetes we propose glaucomaalso to be a type of diabetes. Glaucoma and diabetes share many aspects from various molecular mech-anisms to involvement of insulin and possible use of antidiabetics in glaucoma therapy. Additionally,Alzheimer’s disease has already been proposed to be diabetes type-3. We show that Alzheimer’s diseaseis cerebral glaucoma and diabetes at the same time which, by transitive property of similarities, againleads to our hypothesis that glaucoma is diabetes of the brain. Our proposition may lead to appreciationof certain important facets of glaucoma which have previously not been given due consideration. It alsomay lead to an alternative classification of diabetes as pancreatic and brain diabetes thereby wideningthe vision arena of the understanding of both these disorders.

� 2014 Elsevier Ltd. All rights reserved.

If at first the idea is not absurd then there is no hope for it.Albert Einstein

Hypothesis

We hypothesize that ‘‘Glaucoma is the Diabetes of the Brain’’.Our proposition derives support from our arguments that (i) thereare striking similarities in molecular biology of glaucoma and dia-betes, (ii) brain can also suffer from diabetes, (iii) glaucoma inflictsbrain and the brain part of eye, (iv) insulin is involved in mainte-nance of brain function, intraocular pressure and presumably otherocular functions and (v) brain diabetes is caused by same reagentswhich cause pancreatic diabetes. This hypothesis is important be-cause it may lead to understanding of certain areas of glaucomaand diabetes which have not been evaluated yet. Our hypothesis

is likely to give birth to new areas in glaucoma and diabetes under-standing and management.

Glaucoma–diabetes: a molecular crosstalk

Glaucoma is a progressive ocular neurodegeneration and sec-ond leading cause of blindness with more than 60 million peopleaffected worldwide [1]. The hallmark of glaucoma is gradual lossof retinal ganglion cells (RGCs) [2] by apoptosis [3]. Glaucomahas certain molecular and mechanistic similarities with otherdegenerative disorders of the central nervous system (CNS) likeAlzheimer’s disease (AD) and Parkinson’s disease (PD). Severalpathways for RGC death have been observed in glaucoma [2] andinsulin plays a key role in many of them. Factors implicated inglaucoma pathogenesis include rise in intraocular pressure (IOP),decreased production of neurotrophic factors, excitotoxicity andvasospastic hypoxia [4,5].

RGCs die in glaucoma by various combinations and permuta-tions of apoptosis, oxidative stress (OS), mitochondrial dysfunction,

Page 2: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

536 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

lack of neurotrophic factors, excitotoxicity, insulin deficiency andbrain insulin resistance. Insulin producing b-cells of pancreas(b-cells) in diabetes also suffer demise by apoptosis [6], OS andmitochondrial dysfunction [7], insulin deficiency and insulin resis-tance [8]. There are two pathways for apoptotic death in RGCs viz.intrinsic and extrinsic pathways [9,10]. The same is case withdiabetes where b-cells die by intrinsic [11] and/or extrinsic path-ways [12]. The intrinsic pathways are initiated independent ofany extracellular stimulus. These mainly include cellular stress inwhich apoptotic pathways like c-Jun N-Terminal protein kinases(JNKs) are activated leading to the production of stress associatedprotein kinases (SAPKs) [13,14]. Bcl-2 associated death promoter(BAD) and the other member of same family Bim-EL are also thepro-apoptotic molecules involved [15]. These moieties lead toimpairment in mitochondrial integrity and function which initiatesapoptotic pathways [16] with change in mitochondrial flux andconsequent release of cytochrome-C into the intracellular milieu[16,17]. Our studies also suggest mitochondrial dysfunctionin glaucomatous patients [18,19]. Various cysteine-dependentaspartate-directed proteases (caspases) particularly caspase-3and caspase-9 are then activated to execute cellular death [16].Caspase-3 and caspase-9 mediated apoptosis has been reportedin both RGC death in glaucoma [20] and b-cell death in diabetes[21]. Extrinsic apoptotic pathways have also been implicated inglaucoma and diabetes which involve the binding of certain deathreceptor ligands to various cell surface receptors to bring aboutcellular death [16,17,22] in glaucoma [23] and in diabetes [12].There are many studies which report that inhibition of caspasesenhance the survival of RGCs [24–26]. Blocking caspase-3 leadsto increased survival of RCGs in glaucoma through intrinsic path-way [24–26].

OS has also been shown to be important in RGC death in glau-coma [27] and b-cell death in diabetes [28]. OS mediated RGCdeath in glaucoma comes about either by protein modificationand/or DNA damage [9,29,30] and, interestingly, same is the casewith diabetes [31,32]. This mechanism involves ROS superoxideburst [33]. Additionally, signal transducer and activator of tran-scription-3 (STAT3) pathway regulates neuron survival [34] bydimerization of STAT and its followed translocation to the nucleusfor expression of a battery of genes necessary for RGC survival[35,36]. Activation of the same pathway is the mechanism involvedin b-cell enhanced survival in diabetes [37] also.

Similarities between RGCs and pancreatic b-cells

b-Cells and RGCs have far-reaching similarities in their molecu-lar basis of biological activity. Both cell types show similar geneexpression pattern which includes dopamine b-hydroxylase [38],tyrosine hydroxylase [39], voltage-gated sodium channel type-2[40], glutamic acid decarboxylase [41], glutamate receptor, neuro-filament proteins [42,43], neurotrophin receptors [44,45], thyrot-ropin releasing hormone [46,47], insulin, insulin receptor (IR)[48] and many other genes. This similitude in gene expression pat-tern is indicative of many functional similarities/relations betweenthese two cell types. This may indicate that both RGCs and b-cellsmay be susceptible to similar diseases and may react to certainchallenges in same way; hence bolstering the logic of looking atdiabetes and glaucoma as the same disorder afflicting two sitesof the body. This augments our notion that glaucoma may, infact, be diabetes of the brain. With regards to the similarities inthese molecular aspects, many transcription activators includingIslet-1 [49], Beta-2 [50] and Pax6 [51] are expressed in both the celltypes. In a study performed on various cell lines, control ofexpression of such genes in both neuronal and insulin producingcells was investigated. The investigators evaluated transcriptional

repressor neuron-restrictive silencing factor/repressor elementsilencing transcription factor (NRSF/REST) in the expression of var-ious neuronal genes in insulin-producing cells. NRSF/REST being anegative repressor for neuronal specific genes is present in non-neuronal cell types while as absent in neuronal cells. Theyhave demonstrated that NRSF/REST is absent in insulin and gluca-gon producing cell lines also. Based on their findings, they arguedthat the mechanism of expression of such genes is similar in bothtypes of cells [52] indicating that the molecular impairmentsaffecting these cell types might also be similar. This indicates thatsome identical mechanisms control the molecular biology ofinsulin producing cells and neuronal cells. The b-cells and the neu-ronal cells (including RGCs) also share certain similarities byexpressing many groups of genes like catecholamines, c-aminobutyric acid, certain cell surface receptors intermediary neurofila-ments and many hormones [41,42,44,46,53,54]. Interestinglyb-cells and neurons (including RGCs) are both electrically excitableand are depolarized by exposure to glucose and certain hormones[55,56].

Furthermore, b-cells and neuronal cells express same transcrip-tion factors [57]. A typical example of such a factor is NeuroD/BETA2 [50] which is a basic helix-loop-helix transcription factor in-volved in differentiation to neurons [57] as well as b-cells for trans-activation of insulin gene [50]. Some additional genes that areexpressed by both b-cells and RGCs are trkA and trkC [58,59].Moreover, the home-containing gene paired box protein/Pax6 alsocalled aniridia type-2 protein (AN2) is expressed by both neuronalcells as well as the insulin producing b-cells. Reduced expression ofPax6 transcription factor has been associated with aniridia andglaucoma [60]. On the other hand, mutations in Pax-6 have beenimplicated in glucose intolerance in diabetes [61]. This elucidatesa similitude in molecular pathogenesis of pancreatic diabetes andglaucoma (ocular diabetes).

Insulin signaling in CNS (retina)

Insulin receptor signaling (IRS) impairment has been reportedin many neurological disorders [62]. Interventions that improvethe (CNS) insulin receptor signaling have been employed to treatvarious neurodegenerative diseases which include AD [63]. Insulinsensitizing drugs are, for this reason, an active component of clin-ical trials for AD treatment [64]. By the same rationale, insulin sen-sitizing drugs can be used to treat glaucoma also. The mechanismsof synapse regulation, maintenance and elimination are not com-pletely known but it is mainly the insulin mediated neuronal activ-ity that can be accounted for [65–67]. Synapse maintenance is alsobrought about by insulin like growth factor (IGF)-2 [68–70]. IR; atransmembrane receptor activated by insulin, IGF-1 and IGF-2 isa tyrosine kinase receptor studied extensively for its role in glucosemetabolism. IRs are expressed also in the CNS [71] but their func-tion in CNS has not been sufficiently evaluated though it has beendemonstrated that they are involved in many neuronal functions[72]. IR activates phosphoinositol-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways which control manycellular processes involved in glucose metabolism and cell growthand differentiation [73,74]. One among these functions is theinduction and development of RGCs at the retinal margin [75]. InCNS, IR is expressed in many cell types [71] and its expression levelis developmentally regulated [76]. Maintenance of synaptic plas-ticity and neuronal survival, learning, memory and cellular lifespanmaintenance are also brought about through IR. The importance ofIR in brain circuit development in visual system was recently re-ported in Xenopus tadpoles [76]. The region receiving visual signalsfrom the RGCs (the Xenopus retinotectal system) is important forstudies involving retinal neuron plasticity in vivo. Studies have

Page 3: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546 537

shown that IR is essential for optic tectal neurons to receive normalvisual input from RGCs. IR is, hence, necessary to the normalfunctioning of the visual system which involves RGCs in the visualsystem pathway. A decrease in the expression of IR (a similar con-dition to insulin resistance: as happens in diabetes type-2) leads toa corresponding impairment in visual system functioning [62]. InIR downregulation, the dendritic processes of the tectal neuronsshorten leading to visual impairment [62] presumably mimickingglaucoma. Other studies have reported IR to be very importantfor the functional integrity of the visual system [77–81]. Moreover,reduced IR expression has been demonstrated in many neurode-generative diseases including AD [82] and PD [83]. Since the path-ological characteristics of AD and glaucoma are very similar, it canbe hypothesized that IR downregulation/loss is, at least in part,responsible for glaucomatous vision loss. Amyloid b-plaques(Ab-plaques) are a common finding in glaucoma; IRS preventsthe formation of such plaques [84] and inhibits neurofibrilarytangle formation. IRS also leads to increased degradation ofAb-plaques [85,86]. This finding may play an important role ina number of pathomechanisms of glaucoma. Ab binds with IR, inso doing, decreasing its (insulin’s) effective levels subsequentlycausing OS [87,88] leading to mitochondrial involvement andneuronal cell death. Ab also reduces the IRS by inhibiting Aktactivation. Since Akt pathway is neuroprotective, increased Aband consequently decreased IRS leads to loss of neuroprotectionensuing enhanced neuronal cell death [89]. This is one of the path-ways for RGC loss in glaucoma. Antisense morpholino oligonucleo-tides (AMOs) specifically knockdown IR while preserving the IGF-1receptor. Neurons transfected with AMO suffer from visualresponse insufficiency in vivo. Decrease in IRS by any mechanismdecreases visual responses indicating its significant role invision-neuron function. As ameliorating insulin signaling is beingtried on various neurodegenerative diseases like AD [63,64], weargue, on this premise, glaucoma also is a potential candidate.

In addition to its central role in glucose uptake, insulin playsimportant functional roles in neurons including neuronal growthand maintenance, neuronal degradation by apoptosis, synapticmaintenance and many other CNS centered functions [90]. Gener-ally it is considered that brain is obligatory glucose user uptakingglucose independent of insulin but expression of GLUT-4 in re-sponse to insulin in CNS (particularly RGCs) has been demon-strated [91]. IGF-1 is required for CNS glucose metabolism inmice [92]. Insulin also regulates CNS glucose metabolism in hu-mans [93]. Insulin resistance in CNS is thought to play many rolesin neurodegeneration [94]. The role of insulin resistance in neuro-degenerative disease like AD and glaucoma can be evaluated byinactivation of IR gene in CNS. Markus et al., have reported thedevelopment of brain/neuron-specific insulin receptor knockout(NIRKO) mice model to investigate insulin resistance and neurode-generation [90]. Using this model for conditional inactivation ofIR gene in CNS, they have been able to demonstrate that downreg-ulation of IR in CNS causes impairment in Akt and glycogensynthase kinase-3-beta (GSK-3b) activity and hyperphosphoryla-tion of tau [90]. These impairments are invariably seen in AD andglaucoma pathology though additional mechanisms are alsoinvolved. Insulin resistance in NIRKO mice leads to tau hyper-phosphorylation (typically 3.5-fold) [90], a major part of neurofi-brillary lesions in AD and glaucoma. IRS is important for insulininduced antiapoptotic activity in neurons [90]. Insulin also inhibitsapoptosis in cerebral granule cells [95]. Antiapoptotic activity ofinsulin may be important for many neuronal cell types whichmay include those involved in AD, schizophrenia and glaucoma.On the basis of these findings, it seems obvious that glaucomaand diabetes are mechanistically closely related and may essen-tially be the same molecular conglomeration at two sites of thebody.

Insulin signaling in glaucoma and RGC survival

The brain derived neurotropic factor (BDNF) produced by thetectum [96], leukemia inhibitory factor produced by the astrocytes[97] and IGF-1 produced by astrocytes [98] enhance RGC survival[99]. Insulin and IGF-1 are necessary for RGC survival. Tyrosinephosphorylation of b-subunit of IRs and IGF-1 receptors in culturedchick forebrain neurons and consequently neurite growth andenhancement of neurofilamentous proteins has been reported[100,101]. Insulin also activates protein phosphatase-2-A (PP2A),a serine/threonine phosphatase with broad substrate specificity[102] which regulates the phosphorylation through activation ofGSK-3b [103]. Tau phosphorylation is a vital molecular feature ofglaucoma. The expression of insulin and IGF-1 in human and mon-key RGCs have been confirmed (immunohistochemically) [104].Meyer et al., reported increased RGC survival by insulin and IGF-1 in 8 day old rats [99]. Improvement of RGC survival by insulinis thought to be executed through many pathways [103] includingprotein kinase-C-a (PKCa) activation without translocation [105],PP1 and PP2A activation [106] and maintenance of Bcl2 expressionin stress conditions. Bcl2 activates antioxidant pathways therebylowering free radical formation [107]. Therefore, insulin also en-sures RGC survival by taking care of the OS. Insulin induced Bcl2expression enhances the repair of injured and damaged axons ofRGCs (and those of CNS also) [108]. In a study, exposure of RGCsto insulin enhanced the survival rate by 30% [101]. Insulin inducedBcl2 also ameliorates mitochondrial dysfunction by regulating pro-ton flux [109] and prevents RGC apoptosis.

Avian and mammalian eyes contain neural stem cells and neu-ral stem cells at the retinal margin of post hatch chicken lead todevelopment of certain neurons that integrate into retinal edge[110]. Chick ciliary margin zone (CMZ) cells produce amacrineand bipolar neurons [111]. These cells become restricted to specificcell types with time [112]. It is important to note that the RGCs arenot produced by retinal stem cells in mammals or in birds [75].Keeping that in view, Andy et al., conducted a study about certaingrowth factors that lead to the development of RGCs at the retinalmargin. They interestingly found that insulin leads to the inductionand production of RGCs [75]. RGC development and differentiationby insulin is indicative of some important mechanism by whichinsulin plays the role of an integrative molecule in metabolism ofRGCs in addition to b-cells. It specifies that (a) IRs are continuouslyexpressed in retinal cells, (b) insulin may be important for RGCgrowth and development and (c) insulin signaling is vitally rele-vant to glaucoma. All these factors point to common etiopatho-mechanistic interactomes between glaucoma and diabetes. Thisshows that glaucoma and diabetes are alternative or ectopic formsof each other. Andy and coworkers further reported that insulinalone is sufficient to the proliferative induction of RGCs at retinalmargin [75] giving insulin an important stature in prospectiveglaucoma therapy in addition to diabetes management.

Insulin regulates intraocular pressure (IOP)

IOP elevation leads to optic disc cupping and defects in visualfield ensuing glaucoma [113]. Studies have reported various riskfactors for glaucoma of which IOP is of the greatest importance be-cause of being the only modifiable risk factor [114]. Lowering IOPreduces the risk of glaucoma [115]. Elevated IOP has been shownto be associated with diabetes also [116,117] suggesting someimperative link between glaucoma and diabetes. Moreover, obesityalso can serve as a modifiable risk factor for elevated IOP [118] and,therefore, glaucoma. Leptin might, for this reason, serve as a ther-apeutic agent in glaucoma [20]. IOP elevation has been reported tobe associated with insulin resistance [119] and conversely insulin

Page 4: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

538 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

induced hypoglycemia acutely lowers IOP [120]. These findingsstrongly suggest greater underlying molecular similarities betweenIOP elevation (consequently glaucoma) and diabetes. Many inves-tigators have reported elevation of IOP with elevation in the fastingblood glucose [116,117,121,122] which strongly implies patholog-ical and mechanistic similarities between IOP and glucose metab-olism or, in other words, glaucoma and diabetes. Since insulinregulates the blood glucose levels and insulin deficiency/insulinresistance (diabetes type-1/type-2) is associated with diabetes, itis plausible that insulin deficiency and/or insulin resistance maylead to elevation in IOP [123]. In a recent study conducted on943 human subjects and using HOMA-IR and McAuley index, itwas concluded that there is a strong association between insulinresistance and IOP elevation. In this study, IOP was elevated corre-sponding to the level of insulin resistance [120] even after control-ling the confounding factors [120]. At the current status ofliterature, it can be postulated that insulin activates many signal-ing pathways in retinal neurons many of which play role in cellproliferation, differentiation and expression of various glucosetransporters [91]. Insulin acts in similar ways in pancreatic b-cellsto ward off diabetic pathogenic picture.

Notes from streptozotocin

2-Deoxy-2-[methyl-(nitrosoamino) carbonyl] amino-D-gluco-pyranose (a nitrosamide methylnitrosourea linked to the C2 posi-tion of D-glucose) also called streptozotocin (STZ) is known tocause diabetes mellitus in experimental models. Recently, STZhas been reported to produce biochemical and molecular hall-marks of diabetes mellitus in neurons of CNS. STZ operates throughelevation in OS and consequent DNA damage by release of N-nitr-osoureido mediated production of nitric oxide, superoxides andhydrogen peroxide [124]. As STZ is taken up by the cells that pro-duce insulin, it is taken up by b-cells and also by CNS neurons indi-cating insulin production in CNS. Rat models of cranial diabetesmellitus are produced by intracerebral injection of STZ (ic-STZ).Brain insulin resistance has been established by ic-STZ injections[125]. Reduced cerebral glucose utilization [125], impairment inoxidative metabolism [126], inhibition of IR function [127], cogni-tive deficit and deranged cerebral bioenergetics is a common find-ing post ic-STZ [128]. Many of these impairments occur inglaucoma. In addition to that, the pancreatic cytoarchitecture andinsulin immunoreactivity are same as that in controls in post ic-STZ mice [129]. There are clear regions of brain atrophy in thesemodels in spite of no damage to pancreas. Such models havepathologies typical for glaucoma like glia activation, p53 immuno-reactivity, hyperphosphorylated tau protein and amyloid precursorprotein (APP)-Ab plaques [130,131]. In addition to that, expressionof neurons (Hu), myelin associated glycoprotein-1 (MAG-1) andcholine acetyltransferase (ChAT) and simultaneous elevation inthe expression of p53 [132], tau, APP [130,131], allograft inflam-matory factor-1 (AiF-1) and glial fibrillary acidic protein levels sug-gest an apoptotic mechanism of cell loss in brain diabetes which isquite similar to the cell loss mechanism in glaucoma.

The multifaceted pathological picture of reduced insulin/IGFexpression, impaired insulin/IGF receptor expression, derangementin insulin signaling pathways precipitated by ic-STZ and conse-quent changes in the expression of genes producing a pathologicalpicture mimicking glaucoma (and AD) justify brain diabetes in-duced by ic-STZ independent of pancreatic diabetes. It also sug-gests that insulin sensitizers and insulin supplementation mayameliorate neurodegenerative conditions like glaucoma and AD.Moreover, the molecular derangements that are characteristic ofglaucoma like GSK-3b activation, tau hyperphosphorylation, re-duced survival of neurons can be reproduced by blocking the insu-

lin and its signaling pathways in CNS [133,134]. Molecularnetworks of glaucoma (and diabetes) being intricately interwoven,tau is regulated by insulin and IGF-1 [135] and its ubiquination andphosphorylation is elevated in OS and activation of GSK-3b [136]the way it goes with ic-STZ. APP is also upregulated in neuronalOS [137,138] which explains APP-Ab buildup in both glaucomaas well as ic-STZ treated models.

Pax6 gene involvement in glaucoma and diabetes

Pax6, a highly conserved transcription factor, is the master reg-ulator of eye and retinal development in both invertebrates andvertebrates [139–141]. Mutations in Pax6 which lead to loss offunction present with a ‘no eye’ or ‘small eye’ phenotype [142].Impairment in the Pax6 expression pattern leads to many ocularmalformations which mainly includes developmental defects ofthe anterior chamber of the eye [143–145]. Various enhancers reg-ulate Pax6 expression in neural as well as non-neural ocular tissues[145,146]. Many neural cell types in vertebrate retina express Pax6in different patterns [147].

Pax6 is highly expressed in surface ectoderm and optic cup de-rived cells in mammalian eye development [148]. Optic cup widen-ing (increase in the cup-to-disc ratio) is an important diagnosticfeature of glaucoma. In a pioneering study, Cre/loxP approach wasemployed for selective inactivation of one Pax6 allele in developingdistal optic cup [149]. This relative downregulation of Pax6 in dis-tal optic cup resulted in maldevelopment of iris with no effect inlens [60]. Pax6 insufficiency derived trabecular meshwork (TM)failure leads to elevation in IOP which in turn causes glaucoma[60].

Pax6 is also important in signaling of the development of irido-corneal angle [60]. Obstructions in the TM aqueous outflow and theclosure of iridocorneal angle in Pax6flox/+;LeCre mice has beendemonstrated to lead to almost 25% increase in the basal IOP level.In humans, closure of iridocorneal angle leads to more acute eleva-tion of the IOP where it can raise by a factor of 100% [60] becausethe uveoscleral outflow pathway is not as active in humans, mon-keys and rabbits as it is in mice [150]. IOP elevation operatesthrough axonal damage in optic nerve head (ONH) [151]. Pax6-flox/+;Le-Cre mice develop severe degeneration of axons in the opticnerve similar to other models of glaucoma [152].

It is evident that (a) impairment in aqueous outflow secondaryto developmental defects in TM and (b) iridocorneal angle closurecontribute to juvenile glaucoma in aniridia [60]. Same thing hap-pens in Pax6flox/+;Le-Cre mice viz. TM dysgenesis and closure of iri-docorneal angle caused by iridocorneal adhesions. Both themechanisms cumulatively contribute to optic nerve damage andconsequently glaucoma. Same may be the case in humans also[60].

Pax6 expression has been demonstrated in lens ectoderm, lensplacode, lens vesicle and optic vesicle in the early developmentof mouse ocular system [153,148]. Pax6 has also been reportedin later stages in epithelia of conjunctiva, cornea, lens, ciliary bodyand retinal neurons [154]. The importance of Pax6 in ocular devel-opment can be understood by the finding that the homozygousSmall Eye (Sey) mice harboring mutations in Pax6 gene [142] donot develop eyes and nasal cavities [148,155]. Heterozygous Seymice exhibit smaller eyes with many defects in associated tissues[156]. Heterozygous Pax6 mutations have been implicated in ani-ridia in humans [157] in addition to Peter’s anomaly, certain kera-titis and foveal defects [158]. In our studies, we reported two novelmutations (viz. g.31815391Cytosine > Thymine and g.31823250Thymine > Guanine) in Pax6 gene in cases of ocular malformationamong north-Indians [159]. About 50–75% of patients with aniridiadevelop glaucoma [160] indicating that Pax6 malfunction is

Page 5: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546 539

engaged in glaucomatous phenotype. Aniridia secondary to Pax6mutations leads to elevation of IOP [160,161].

The pathway for glaucoma pathogenesis secondary to Pax6mutation derived aniridia is not completely understood but it isthought that anterior chamber dysgenesis plays an important role.Malformations in the anterior chamber lead to impaired aqueousoutflow which leads to elevation in IOP [162]. Abnormal differen-tiation of TM and absence of Schlem’s canal have been reportedin aniridia on histopathological examinations [163]. This suggeststhat TM dysgenesis can also be due to Pax6 mutations ensuingglaucoma. However, anterior chamber dysgenesis has been indi-cated in other gene defects also like Pitx2, Pitx3, Foxc1, Foxe3,Lmx1b, Maf. It is interesting to note that Pax6 controls the expres-sion of many transcription factors which includes Maf [164].

In an important study Daniela et al., examined the role of Pax6in anterior segment development in heterozygous Pax6LacZ+ defi-cient mice [165] and found that Pax6 haplo-insufficiency is respon-sible for impaired anterior chamber mesenchyme differentiation,TM dysmorphogenesis and maldevelopment of Schlemm’s canal[166]. All these are important factors for development of varioustypes of glaucoma. In a case study involving two children with ani-ridia and Wilm’s tumor, deletion of short arm of chromosome-11was reported [167]. Pax6 is located on chromosome-11 short arm.

TM is embryologically derived from neural crest [168]. Pax6 isimportant for the cranial neural crest derived tissue morphogene-sis as shown in Sey rats where the migration of neural crest cellsfrom mid brain to craniofacial locations is hampered [169]. Pax6insufficiency is also involved in defects in many other neural crestderived ocular tissues [156]. Pax6 expression has been reported inoptic cup neuroectoderm derived ocular cells also [153,148,154].Iris and eye size impairments are not there in Pax6+/+-Pax6+/� chi-meric mice [170].

Lot of work has been done in understanding the mechanismsthat maintain and regulate insulin producing b-cell function[171,172]. Pax6 plays very important role in the development ofocular tissues and pancreas [173] and also CNS. Heterozygous lossof function mutations in Pax6 in mammals lead to many ocularabnormalities while the homozygotes are born without eyes aswell as pancreatic malformations and die at birth [142,174]. Thissuggests that Pax6 seems to be intricately involved in both pancre-atic diabetes and ocular diabetes (glaucoma). Development andspatial organization of islet cell precursors require Pax6[174,175]. Pax6 is regulated by the transcription factor pancreaticand duodenal homebox-1/insulin factor promoter-1 (Pdx1) [176].Following the development of the pancreatic cytoarchitecture,Pax6 controls the expression of insulin, glucagon, somatostatinand pancreatic polypeptide [175,177].

Since many developmentally active gene regulating moietiescontinue to be active in later stages also [172,178]; Pax6 being ex-pressed in both development of eyes and pancreas and then per-sisting in both the tissues in adult stages [179–181] suggests thatit has key roles to play in the maintenance of ocular and pancreatictissue functional integrity. In addition to predisposition to glau-coma through aniridia and IOP elevation, heterozygous Pax6 muta-tions have been reported to predispose humans to diabetes also[179–182].

In a study carried on conditional Pax6 mouse mutants (tamox-ifen inducible Cre deletion strain), it was demonstrated that assoon as the loss of functional Pax6 occurs, diabetes symptoms oc-cur with acute penetrance [183]. The investigators also reportedsevere impairment in production of many pancreatic hormones,transcription factors and insulin processing enzymes with Pax6functional deficit in these mice indicating Pax6 is essential to b-cellfunction and maintenance of pancreatic functionality [183].

Role of Pax6 in insulin production has been studied [175,177],regulatory regions described [175,177,184] and expression of

Pax6 in b-cells confirmed [179,180]; lending support to the obser-vation that glucose regulation and insulin kinetics is deranged inindividuals with heterozygous Pax6 mutations [179,180,185].Pax6 acts upstream of Pdx1 and Nkx6.1; owing to their downreg-ulation with Pax6 loss of function in adults. On the other hand,Pdx1 acts upstream of Pax6 during embryonic development (a typ-ical example of cross regulated reciprocal feed loop) [183]. Thisindicates that Pdx1 and Pax6 are very intricately connected forthe development and maintenance of pancreas. Correlatively,Pax6 and Pdx1 have been reported to be colocalized with glu-cose-dependent insulinotropic polypeptide (GIP) in mouse retinalcells [186] indicating their synergistic function in developmentand maintenance of retinal cells also.

Hyperinsulinemia in diabetes type-2 is caused by impairments inproinsulin processing with prohormone convertase (PC) 1/3 and PC2and carboxypeptidase E [187]. PC1 and PC3 are drastically downreg-ulated in mice with Pax6 null mutations [188] meaning that Pax6affects insulin processing showground either directly or throughPdx1. Patients with aniridia secondary to Pax6 mutations harborhigher levels of proinsulin in their prediabetic states [183].

Similarities between glaucoma and Alzheimer’s disease

Glaucoma and AD share many characteristics [189,190] on thebasis of which it has been postulated that glaucoma and AD isthe same disease affecting different parts of the brain. Glaucoma,for this reason, has been referred to as ‘‘Ocular Alzheimer’s Dis-ease’’ [191]. Both eye and optic nerve develop embryologicallyfrom the third ventricle [192]. Intraocular and intracranial spaceshave similar ranges for physiological pressure and also respondin a similar manner to intra-abdominal and intra-thoracic pressuredynamics [193]. IOP generation and maintenance is dependent onthe rate of production and outflow of aqueous humor in the ante-rior chamber of the eye [194,195]. The path followed by the aque-ous fluid is production by ciliary body epithelium, passing throughthe pupil and exiting through TM just before entering the systemiccirculation through Schlem’s canal. Similarly, intracranial pressure(ICP) is generated and maintained through production by choroidplexus (neuroepitheluim), passage through brain ventricles andreabsorption of CSF into the lymphatic system [194]. Since, retinais an extension of brain and glaucoma involves degeneration ofRGC (which are neurons), glaucoma is typically seen as a neurode-generative disorder. For this reason both glaucoma and AD areslowly progressing age related neurodegenerative disorders[196,197]. One of the pathological pictures for AD is decreased le-vel of Ab [198] and increased levels of tau in CSF [199]. Decreasedlevels of Ab and increased levels of tau have been reported also inthe vitreous fluid of glaucoma and diabetic retinopathy patients[200]. Ab-plaque accumulation has been reported in RGCs of ratmodels of glaucoma. Both glaucoma and AD involve activation ofcaspases in their pathogenesis. It is the caspase activation and con-sequent APP misprocessing that leads to Ab-plaques in AD [201]while it is the same thing that happens in RGCs in glaucoma. Glau-comatous death of RGCs involves activation of caspase-3 which issimilar to that of the apoptotic mechanism of b-cells in diabetes[202]. Based on these findings, it is thought that pathogenic mech-anisms of glaucoma are same as that of AD [191]. The way IOP ele-vation occurs in glaucoma, ICP is presumably elevated in AD [203].And there are cases of normotensive glaucoma [204] the way thereare normotensive AD cases.

Similar pathology of glaucoma, Alzheimer’s and diabetes

AD shares a great deal of biochemical, pathological and mecha-nistic abnormalities with diabetes [205,206] and glaucoma [129].

Page 6: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

540 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

AD and glaucoma present with many common characteristicsincluding increased cell death, abundant neurofibrillary tangles,consequent dystrophic neuritis, impaired deposits of APP, Ab-pla-ques, pTau protein deposition, increased expression of apoptoticgenes with corresponding increase in relevant signal transductionpathways, deranged energy metabolism and mitochondrial dys-function with elevated OS causing DNA damage. These two dis-eases are substantially similar in many respects and may,therefore, be the same disease finding expression at two differentlocations of the same organ (i.e. brain). This notion can be justifiedif the pathogenic mechanisms of both the disorders are strikinglysimilar. A coherent pathological picture that explains all these phe-nomena is likely to describe many aspects of these two disordersand may pave way for development of novel and effective thera-pies [137]. Insulin signaling dysfunction explains many aspects ofsuch a picture. Insulin/IGF-1 bind to corresponding receptors onthe cell surface and activate a plethora of intricately connected sig-nal transduction pathways including autophosphorylation of tyro-sine kinases [207,208] in consequence phosphorylating IRs[207,209]. The process activates extracellular signal-related ki-nase/mitogen-activated protein kinase (ERK/MAPK) and PI3K/Akt(protein kinase B) pathways inhibiting GSK-3b. These pathwaysare involved in inhibition of apoptosis, cell growth (and prolifera-tion) and reduction in OS [209–213]. Chronic brain insulin resis-tance and insulin deficiency has been reported in AD[137,138,209,214] and improvement in cognitive function by useof insulin sensitizers and intranasal insulin has also been reported[130,215,216]. Conversely cognitive impairments have been testi-fied in animal models of type-2 diabetes [217,218].

There is significantly lower number of RGCs in optic nerves ofAD cadavers as compared to controls [219]. Glaucoma patientswith elevated IOP present with optic nerve characteristics similarto those of AD which includes thinning of retinal nerve fiber layer(RNFL), increased optic cupping and abnormal electroretinogram[220]. It is interesting to note that loss of vision begins with lossof axons in mid brain and then the damage progresses anteriorlyengulfing optic nerve and the fundoscopically visible area in mouse[221] and primate models of glaucoma [222,223]. AD diagnosis isdifficult owing to expensive neuroimaging methods and invasivesampling of the CSF. Glaucoma is the most common retinal disor-der that has been matched up to AD [224].

Though glaucoma is idiopathic, ONH cupping, nerve fiber layer(NFL) thinning and activation of glia are commonly seen leading toRGC death and consequent vision loss [225] thereby putting glau-coma into the class of true neurodegenerative disorders. This is fur-ther supported by the fact that transynaptic degeneration has beenshown in lateral geniculate nucleus in visual cortex of glaucomapatients [226,227]. Though IOP is the most common modifiablefactor in glaucoma [228], but IOP reduction does not reverseONH cupping and retinal degeneration and, therefore, almost allpatients develop optic neuropathy [225,229]. In addition, manyopen angle glaucoma patients have IOP within the normal range[230]. Increased prevalence of glaucoma in AD is now well known[231–233]. In a study 25.9% of AD patients also presented withglaucoma as compared to 5.2% in control group [231]. As is the casewith AD, glaucoma patients also present with impaired congnitivefunctions [232] although there are studies that do not support this[233].

Glaucoma and AD having strikingly common pathologies [234];the retinal regions that are affected in AD are same as those af-fected in glaucoma viz. RGC death [235] and NFL thinning in ADand RGC death, NFL thinning and optic nerve hypoplasia in glau-coma [235–237]. Like glaucoma, glial activation secondary toRGC loss has also been demonstrated in AD [238]. The retinalhemodynamic parameters of AD patients also are similar to thoseof glaucoma patients with both featuring narrowing of vessels

and reduced blood flow [239–241] which leads to increase in theactivity of Hypoxia-inducible factors (HIFs) [242]. pTau has beenreported in the inner retina [243], optic nerve and peripappilaryglia [244] of glaucoma patients [245]. pTau has also been impli-cated in neurofibrillary threads and tangles (NFT) formation inAD. Reddy et al., reported decrease in Ab in AD [246]. Retinal gliais activated in both AD [247] and glaucoma [234].

Alzheimer’s disease as type-3 and glaucoma as type-4 diabetes

In one of our previous studies we provided strong rationale toestablish a link between diabetes and encephalization [248] fromevolutionary point of view. We argued that diabetes has evolution-ary links with neurons and brain activity in humans. Our studiesare also supported by studies inspired by human evolution andanthropology [249]. Impaired cerebral glucose utilization and de-ranged bioenergetics in AD is precipitated early in the develop-ment of disease [250–252] and same is the case with glaucoma[253,254]. Owing to many similarities AD has recently been pro-posed to be another form of diabetes representing ‘‘type-3 diabe-tes’’ [129,137]. By the same token, glaucoma can also beproposed to be another type of diabetes, may be ‘‘diabetes type-4’’.

Irrespective of the types, diabetes is, for the most part, derange-ment in insulin functioning machinery precipitating hyperglyce-mia. Type-2 is the most common form of diabetes mellitus and ismediated by insulin resistance as a consequence of reduction inIR expression, impairment in IR-tyrosine kinase functioning, insu-lin receptor substrate type-1 expression [255]. AD shows up withthe characteristics of both type-1 and type-2 diabetes by present-ing insulin resistance as well as hypoinsulinemia restricted to theCNS [129]. Malfunction of the insulin machinery causes cognitivedysfunction in AD [137,138] explained by the fact that expressionand phosphorylation of tau gene and its protein product respec-tively are regulated by insulin and IGF [256,257]. Studies havedemonstrated that many hallmarks of neurodegeneration thatare seen in AD can be reproduced by mutilation of insulin path-ways [258–260].

By studying the post mortem tissues of advanced AD patients,Suzanne et al., showed that AD type neurodegeneration is hand-cuffed with significant aberrations in expression of insulin andgenes involved in its downstream pathways [129,130,137]. Highlyreduced levels of insulin and IGF-1 and their receptor genes wasreported by these authors in advanced cases of AD. They arguedthat the signaling pathways that are activated by insulin andIGF-1 (which are mediators for neuronal viability, tau expression,neuronal bioenergetics and mitochondrial function) were de-ranged in their subjects. The Suzanne et al., study is important inthe sense that they showed insulin to be linked with neurodegen-eration (AD in particular); owing to the striking similarities seen inAD brain (in patients without any diabetes) and diabetes (bothtypes) which included impairments in IGFs [129,130,261] thatare indispensible for b-cell function [262,263]. On the rationaleof the above discussions, it can be speculated that AD and diabetesis the same disease with manifestation in two different organs.Therefore, the term ‘‘diabetes type-3’’ for AD has found its placein literature. This paper is centered upon providing conceptual ba-sis of the idea that same could be the case with glaucoma and it isapt to think of glaucoma as diabetes type-4.

Brains at different severity stages (Braak stages) of AD havebeen studied to test if the extent of derangement in insulin/IGFdeficiency in the brain corresponds with the severity of the disor-der [264,265]. In these studies insulin, IGF-1, IGF-2, IR, tau, APPexpression was shown to be regulated in a way that correspondswith the Braak staging of AD. This indicates that insulin mediatedpathogenesis underlies the mechanism of AD [138]. These studies

Page 7: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546 541

demonstrated that impairments in insulin, IGF signaling and theirdownstream processes lie in the genesis of AD and are immenselyimportant to understand the disease.

Neuronal and oligodenroglial cell viability depends on the prop-er functioning of insulin and IGF interactome in the brain[138,130,131,260,266–268], derangements which lead to impair-ment in neuronal bioenergetics, mitochondrial dysfunction, proin-flammatory cytokine expression, activation of microglia andrelated neuronal inflammation [130,138,268] and APP expressionin early stages of AD [269] which increases OS [136]. Same pro-cesses are activated in glaucoma pathogenesis also. It must benoted that microglia activation and increased expression of astro-cytic genes lead to further increase in CNS insulin/IGF-deficiency/resistance in AD [129]. Activation of microglia leads to APP-Abbuildup [269–273]. The diabetes type-3 based mechanism of ADseems to be mediated through impaired insulin/IGF signalingwhich increases OS [127,128,259] mediated by increased APPexpression and cleavage [136]. Elevated APP expression and Abbuildup form adducts which are neurotoxic [273–275]. These ad-ducts are seen in glaucoma also and may contribute to RGC death.

There is also an insulin mediated mechanism for cytoskeletalcollapse in AD. Braak stage dependent decreased expression inChAT and its reduced colocalization with insulin/IGF-1 receptorsuggest that neuronal ChAT is regulated by insulin/IGF-1 [267]. De-cline in ChAT and tau functionalities lead to cytoskeletal collapse,loss of neuronal plasticity, synaptic degeneration and decreasedproduction of neurotransmitter acetylcholine. Insulin/IGF, there-fore, are at the center-stage of AD pathology and, hence, AD canbe thought of another form of diabetes [129].

Glaucoma and antidiabetic drugs

Insulin/IGF deficiency, insulin resistance very early in glaucomapathogenesis and subsequent derangements in insulin signalingpathways and downstream processes [137,138]; in addition tothe fact that brain diabetes can be caused by ic-STZ indicate thatglaucomatous and AD neurodegeneration could be prevented or,at least, delayed by intracerebral delivery of insulin, insulin-sensi-tizing drugs like peroxisome proliferator-activated receptor (PPAR)agonists (a group of nuclear receptor isoforms). Through their tran-scription activity, they lead to the activation of insulin responsiveelements and allied signaling pathways. All the three isoforms ofPPAR viz. PPAR-a, PPAR-c and PPAR-d are expressed in adult brainin humans [269]. In an experiment involving ic-STZ treated rats,the effect of low doses of PPAR (PPAR-a: 25 lg/kg, PPAR-d: 2 lg/kg,or PPAR-c: 20 lg/kg) isoforms was evaluated with respect toprevention of neurodegeneration, rescue of insulin/IGF responsivecells [130]. PPAR agonists presumably rescued insulin responsivecells (neurons and oligodendria) [129]. Additionally, there is an in-crease in the insulin receptor expression and insulin binding in re-sponse to PPAR treatment in ic-STZ treated rats [129]. This may bethe raison d’être for further studies involving insulin sensitizers fortreatment of glaucoma. PPAR agonists bring about increasedexpression of ChAT by rescuing ChAT cells. ChAT cells are abundantin retina which includes ganglion cells [276] with the abundance of11.5% in the ganglion cell layer [277]. Additionally, minocycline (anantibiotic) is a neuroprotective agent which has been reported toameliorate neurodegeneration in both AD models [278–280] andRGC loss in glaucoma [281–283].

How to test the hypothesis?

Insulin influences a number of central nervous functions inaddition to maintenance of blood glucose levels. If glaucoma is dia-betes of the brain then insulin and other related antidiabetics

should be able to ameliorate glaucomatous conditions. A studycould be designed where glaucomatous animal models (geneticsand other confounding factors ruled out) are treated with insulindelivered specifically to the brain. The two main problems in sucha study are brain specific delivery of insulin and safety of such aninsulin administration. Delivery of insulin exclusively to brain non-invasively has not been achieved to the best of our knowledge.However, partial brain specific insulin delivery can be executedby either intracerebroventricular administration of insulin oradministration of insulin intranasaly. After administration intran-asaly, insulin enters the CSF from nasal mucosa through bulbusolfactorius and intercellular clefts [284]. The improvement in glau-coma symptoms in subjects as against controls will partially con-firm the hypothesis. However, there will be a number ofconfounding factors which need to be ruled out. The safety of intra-nasal delivery of insulin to otherwise non-diabetic glaucomatousindividuals needs to be worked out before clinical trials areexecuted.

Discussion

In this paper, we discuss the molecular biology that is strikinglysimilar between glaucoma and diabetes. The molecular biology andetiopathogenesis of glaucoma and diabetes share a number ofimportant features. RGCs are the cells that die in glaucoma whileb-cell death stands the hallmark of diabetes. RGCs and pancreaticb-cells are similar in many respects important in the etiopathogen-ic and mechanistic picture of their respective diseases. Literaturesuggests that insulin signaling is important in CNS and RGC growthand differentiation. IR and insulin signaling is important in thedevelopment and maintenance of visual system. Insulin also hasantiapoptotic activities for RGCs. To add to this, insulin regulatesthe IOP in addition to the regulation of blood glucose concentra-tion. The way STZ causes diabetes in experimental models; ic-STZ causes brain diabetes like condition. This means that there issomething called brain diabetes which can be precipitated by thesame moiety used to establish pancreatic diabetes. This is animportant etiological handcuff between CNS neurodegeneration(including glaucoma) and pancreatic diabetes. Pax6 gene is veryimportant in the development and maintenance of eyes across allvertebrates. At the same time, it is now well established thatPax6 gene is similarly important in the development and mainte-nance of b-cells. Since AD has striking similarities with glaucomaas well as diabetes (AD has been proposed to be diabetes type-3),it is natural to think of glaucoma as a type of diabetes. This discus-sion indicates that diabetes and glaucoma may essentially be thesame disease with manifestation at different sites of the body. Thatwill help us in understanding glaucoma from a different perspec-tive which may improve our understanding of its etiological, mech-anistic and therapeutic aspects. We also put emphasis on thepotential use of antidiabetic drugs in treatment of glaucoma.

Conflict of interest

All the authors declare ‘‘No conflict of interest’’.

Acknowledgement

Muneeb Faiq thankfully acknowledges Senior Research Fellow-ship from Indian Council of Medical Research.

References

[1] Quigley HA, Broman A. The number of persons with glaucoma worldwide in2010 and 2020. Br J Ophthalmol 2006;90:151–6.

[2] Qu J, Wang D, Grosskreutz CL. Mechanisms of retinal ganglion cell injury anddefense in glaucoma. Exp Eye Res 2010;91(1):48–53.

Page 8: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

542 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

[3] Okisaka S, Murakami A, Mizukawa A, Ito J. Apoptosis in retinal ganglioncell decrease in human glaucomatous eyes. Jpn J Ophthalmol 1997;41:84–8.

[4] Kuehn MH, Fingert JH, Kwon YH. Retinal ganglion cell death in glaucoma:mechanisms and neuroprotective strategies. Ophthalmol Clin North Am2005;18(3):383–95.

[5] Yamamoto T. The dawn of neuroprotective therapy for glaucomatous opticneuropathy. Nihon Ganka Gakkai Zasshi 2001;105(12):866–83.

[6] Johnson JD, Luciani DS. Mechanisms of pancreatic beta-cell apoptosis indiabetes and its therapies. Adv Exp Med Biol 2010;654:447–62.

[7] Donath MY, Ehses JA, Maedler K, Schumann DM, Ellingsgaard H, Eppler E,et al. Mechanisms of beta-cell death in type 2 diabetes. Diabetes2005;54(Suppl 2):108–13.

[8] Salsali A, Nathan M. A review of types 1 and 2 diabetes mellitus and theirtreatment with insulin. Am J Ther 2006;13(4):349–61.

[9] Almasieh M, Wilson AM, Morquette B, Cueva Vargas JL, Di Polo A. Themolecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res2012;31(2):152–81.

[10] Gariano RF, Gardner TW. Retinal angiogenesis in development and disease.Nature 2005;438(7070):960–6.

[11] Grunnet LG, Aikin R, Tonnesen MF, Paraskevas S, Blaabierg L, Storling J, et al.Proinflammatory cytokines activate the intrinsic apoptotic pathway in b-cells. Diabetes 2009;58(8):1807–15.

[12] De Franco S, Chiocchetti A, Ferretti M, Castelli L, Cadario F, Cerutti F, et al.Defective function of the Fas apoptotic pathway in type 1 diabetes mellituscorrelates with age at onset. Int J Immunopathol Pharmacol2007;20(3):567–76.

[13] Brecht S, Kirchhof R, Chromik A, Willesen M, Nicolaus T, Raivich G, et al.Specific pathophysiological functions of JNK isoforms in the brain. Eur JNeurosci 2005;21(2):363–77.

[14] Tezel G, Chauhan BC, LeBlanc RP, Wax MB. Immunohistochemical assessmentof the glial mitogen-activated protein kinase activation in glaucoma. InvestOphthalmol Vis Sci 2003;44(7):3025–33.

[15] Wiggins CM, Tsvetkov P, Johnson M, Joyce CL, Lamb CA, Bryant NJ, et al.BIM(EL), an intrinsically disordered protein, is degraded by 20S proteasomesin the absence of polyubiquitylation. J Cell Sci 2011;124(6):969–77.

[16] Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol2007;35(4):495–516.

[17] Ashkenazi A, Holland P, Eckhardt SG. Ligand-based targeting of apoptosis incancer: the potential of recombinant human apoptosis ligand 2/Tumornecrosis factor-related apoptosis-inducing ligand (rhApo2L/TRAIL). J ClinOncol 2008;26(21):3621–30.

[18] Kumar M, Tanwar M, Faiq MA, Pani J, Shamsi MB, Dada T, et al. MitochondrialDNA nucleotide changes in primary congenital glaucoma patients. Mol Vis2013;19:220–30.

[19] Tanwar M, Dada T, Sihota R, Dada R. Mitochondrial DNA analysis in primarycongenital glaucoma. Mol Vis 2010;16:518–33.

[20] Gupta A. Leptin as a neuroprotective agent in glaucoma. Med Hypoth2012;81(5):797–802.

[21] Liadis N, Murakami K, Eweida M, Elford AR, Sheu L, Gaisano HY, et al.Caspase-3-dependent b-cell apoptosis in the initiation of autoimmunediabetes mellitus. Mol Cell Biol 2005;25(9):3620–9.

[22] Bazzoni F, Beutler B. The tumor necrosis factor ligand and receptor families. NEngl J Med 1996;334(26):1717–25.

[23] Lebrun-Julien F, Bertrand MJ, De Backer O, De Backer O, Stellwagen D,Morales CR, et al. ProNGF induces TNFalpha-dependent death of retinalganglion cells through a p75NTR non-cellautonomous signaling pathway.Proc Natl Acad Sci USA 2010;107(8):3817–22.

[24] Chaudhary P, Ahmed F, Quebada P, Sharma SC. Caspase inhibitors block theretinal ganglion cell death following optic nerve transection. Mol Brain Res1999;67:36–45.

[25] Kermer P, Ankerhold R, Klöcker N, Krajewski S, Reed JC, Bähr M. Caspase-9:involvement in secondary death of axotomized rat retinal ganglion cellsin vivo. Mol Brain Res 2000;85:144–50.

[26] Kermer P, Klocker N, Labes M, Bahr M. Inhibition of CPP32-like proteasesrescues axotomized retinal ganglion cells from secondary cell death in vivo. JNeurosci 1998;18:4656–62.

[27] Schlieve CR, Tam A, Nilsson BL, Lieven CJ, Raines RT, Levin LA. Synthesis andcharacterization of a novel class of reducing agents that are highlyneuroprotective for retinal ganglion cells. Exp Eye Res 2006;83:1252–9.

[28] Maritim AC, Sanders RA, Watkins JB. Diabetes, oxidative stress, andantioxidants: a review. J Biochem Mol Toxicol 2003;17(1):24–38.

[29] Cross JV, Templeton DJ. Thiol oxidation of cell signaling proteins: controllingan apoptotic equilibrium. J Cell Biochem 2004;93(1):104–11.

[30] Izzotti A, Bagnis A, Saccã SC. The role of oxidative stress in glaucoma. MutatRes 2006;612(2):105–14.

[31] Baynes JW, Thorpe SR. Role of oxidative stress in diabetic complications: anew perspective on an old paradigm. Diabetes 1999;48(1):1–9.

[32] Blasiak J, Arabski M, Krupa R, Wozniak K, Zadrozny M, Kasznicki J, et al. DNAdamage and repair in type 2 diabetes mellitus. Mutat Res 2004;554(1–2):297–304.

[33] Nguyen SM, Alexejun CN, Levin LA. Amplification of a reactive oxygen speciessignal in axotomized retinal ganglion cells. Antioxid Redox Signal2003;5(5):629–34.

[34] Huang Y, Li Z, Wang N, van Rooijen N, Cui Q. Roles of PI3K and JAK pathwaysin viability of retinal ganglion cells after acute elevation of intraocular

pressure in rats with different autoimmune backgrounds. BMC Neurosci2008;11(9):78.

[35] Signore AP, Zhang F, Weng Z, Gao Y, Chen J. Leptin neuroprotection in theCNS: mechanisms and therapeutic potentials. J Neurochem2008;106(5):1977–90.

[36] Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identification of SOCS-3 as a potential mediator of central leptin resistance. Mol Cell1998;1:619–25.

[37] Hoang PT, Park P, Cobb LJ, Paharkova-Vatchkova V, Hakimi M, Cohen P, et al.The neurosurvival factor Humanin inhibits beta cell apoptosis via Stat3activation and delays and ameliorates diabetes in NOD mice. Metabolism2010;59(3):343–9.

[38] Watanabe T, Nagatsu I. Immunohistochemical colocalization of insulin,aromatic L-amino acid decarboxylase and dopamine beta-hydroxylase inislet B cell chicken pancreas. Cell Tissue Res 1991;263:131–6.

[39] Teitelman G, Lee JK. Cell lineage analysis of pancreatic islet development:glucagon and insulin cells arise from catecholaminergic precursors present inthe pancreatic duct. Dev Biol 1987;121:454–66.

[40] Philipson LH, Kusnetsov A, Larson T, Zeng Y, Westermark G. Diabetes1993;42:1372–7.

[41] Baekkeskov S, Aanstoot HJ, Christgau S, Reetz A, Solimena M, Cascalho M,et al. Nature 1990;347:151–7.

[42] Teitelman G. Dev Biol 1990;142:368–79.[43] Atouf F, Tazi A, Polak M, Czernichow P, Scharfmann R. J Neuroendocrinol

1995;7:957–64.[44] Scharfmann R, Tazi A, Polak M, Kanaka C, Czernichow P. Diabetes

1993;42:1829–36.[45] Polak M, Scharfmann R, Seilheimer BEG, Dressler D, Verma IM, Potter H. Proc

Natl Acad Sci USA 1993;90:5781–5.[46] Martino E, Seo H, Lernmark A, Refetoff S. Proc Natl Acad Sci USA

1980;77:4345–8.[47] Scharfmann R, Leduque P, Aratan Spire S, Dubois P, Basmaciogullari A,

Czernichow P. Endocrinology 1988;123:1329–33.[48] Holdengreber V, Ren Y, Ben-Shaul Y, Hausman RE. Co-localization of the

insulin receptor, jun protein and choline acetyltransferase in embryonic chickretina. Exp Eye Res 1998;66(3):307–13.

[49] Karlsson O, Thor S, Norbert T, Ohlsson H, Edlund T. Nature 1990;344:879–82.[50] Naya FJ, Stellrecht CMM, Tsai M-J. Genes Dev 1995;9:1009–19.[51] Turque N, Plaza S, Radvanyi F, Carriere C, Saule S. Mol Endocrinol

1994;8:929–38.[52] Atouf F, Czernichow P, Scharfmann R. Expression of neuronal traits in

pancreatic beta cells implications of neuron-restrictive silencing factor/repressor element silencing transcription factor, a neuron-restrictive silencer.J Biol Chem 1997;272(3):1929–34.

[53] Tazi A, Le Bras S, Lamghitnia HO, Vincent JD, Czernichow P, Scharfmann R. JBiol Chem 1996;271:10154–60.

[54] Zhang B, van Adel BA, Gabriele J, Duong M, Henry P, Ball AK. Expression of the40 kDa catecholamine regulated protein in the normal and injured rat retina.J Chem Neuroanat 2002;24(1):41–8.

[55] Russell JW, Golovoy D, Vincent AM, Mahendru PIA, Olzmann JA, Mentzer A,et al. High glucose-induced oxidative stress and mitochondrial dysfunction inneurons. FASEB 2002;16(13):1738–48.

[56] Santiago AR, Rosa SC, Santos PF, Cristóvão AJ, Barber AJ, Ambrósio AF.Elevated glucose changes the expression of ionotropic glutamate receptorsubunits and impairs calcium homeostasis in retinal neural cells. InvestOphthalmol Vis Sci 2006;47(9):4130–7.

[57] Lee JE, Hollenberg S, Snider L, Turner DL, Lipnick N, Weintraub H. Conversionof Xenopus ectoderm into neurons by NeuroD, a basic helix-loop-helixprotein. Science 1995;268:836–44.

[58] Cui Q, Tang LS, Hu B, So KF, Yip HK. Expression of trkA, trkB, and trkC ininjured and regenerating retinal ganglion cells of adult rats. InvestOphthalmol Vis Sci 2002;43(6):1954–64.

[59] Miralles F, Philippe P, Czernichow P, Scharfmann R. Expression of nervegrowth factor and its high-affinity receptor Trk-A in the rat pancreas duringembryonic and fetal life. J Endocrinol 1998;156(3):431–9.

[60] Kroeber M, Davis N, Holzmann S, Kritzenberger M, Shelah-Goraly M, Ofri R,et al. Reduced expression of Pax6 in lens and cornea of mutant mice leads tofailure of chamber angle development and juvenile glaucoma. Hum MolGenet 2010;19(17):3332–42.

[61] Yasuda T, Kajimoto Y, Fujitani Y, Watada H, Yamamoto S, Watarai T, et al.PAX6 mutation as a genetic factor common to aniridia and glucoseintolerance. Diabetes 2002;51(1):224–30.

[62] Chiu SL, Cline HT. Insulin receptor signaling in the development of neuronalstructure and function. Neural Dev 2010;5:8104–7.

[63] Landreth G. Therapeutic use of agonists of the nuclear receptor PPARgammain Alzheimer’s disease. Curr Alzheimer Res 2007;4:159–64.

[64] Watson GS, Craft S. The role of insulin resistance in the pathogenesis ofAlzheimer’s disease: implications for treatment. CNS Drugs 2003;17:27–45.

[65] Hashimoto K, Kano M. Functional differentiation of multiple climbing fiberinputs during synapse elimination in the developing cerebellum. Neuron2003;38:785–96.

[66] Colman H, Nabekura J, Lichtman JW. Alterations in synaptic strengthpreceding axon withdrawal. Science 1997;275:356–61.

[67] Bosman LW, Takechi H, Hartmann J, Eilers J, Konnerth A. Homosynaptic long-term synaptic potentiation of the ‘‘winner’’ climbing fiber synapse indeveloping Purkinje cells. J Neurosci 2008;28:798–807.

Page 9: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546 543

[68] Huh GS, Boulanger LM, Du H, Riquelme PA, Brotz TM, Shatz CJ. Functionalrequirement for class I MHC in CNS development and plasticity. Science2000;290:2155–9.

[69] Ishii DN. Relationship of insulin-like growth factor II gene expression inmuscle to synaptogenesis. Proc Natl Acad Sci USA 1989;86:2898–902.

[70] Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al.The classical complement cascade mediates CNS synapse elimination. Cell2007;131:1164–78.

[71] Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributedin the central nervous system of the rat. Nature 1978;272:827–9.

[72] Choi J, Ko J, Racz B, Burette A, Lee JR, Kim S, et al. Regulation of dendritic spinemorphogenesis by insulin receptor substrate 53, a downstream effector ofRac1 and Cdc42 small GTPases. J Neurosci 2005;25:869–79.

[73] Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipidmetabolism. Nature 2001;414:799–806.

[74] Schulingkamp RJ, Pagano TC, Hung D, Raffa RB. Insulin receptors and insulinaction in the brain: review and clinical implications. Neurosci Biobehav Rev2000;24:855–72.

[75] Fischer AJ, Dierks BD, Reh TA. Exogenous growth factors induce theproduction of ganglion cells at the retinal margin. Development2002;129(9):2283–91.

[76] Chiu SL, Chen CM, Cline HT. Insulin receptor signaling regulates synapsenumber, dendritic plasticity, and circuit function in vivo. Neuron2008;58:708–19.

[77] Aizenman CD, Cline HT. Enhanced visual activity in vivo forms nascentsynapses in the developing retinotectal projection. J Neurophysiol2007;97:2949–57.

[78] Sin WC, Haas K, Ruthazer ES, Cline HT. Dendrite growth increased by visualactivity requires NMDA receptor and Rho GTPases. Nature 2002;419:475–80.

[79] Shen W, Da Silva JS, He H, Cline HT. Type A GABA-receptor-dependentsynaptic transmission sculpts dendritic arbor structure in Xenopus tadpolesin vivo. J Neurosci 2009;29:5032–43.

[80] Zhang LI, Tao HW, Poo M. Visual input induces long-term potentiation ofdeveloping retinotectal synapses. Nat Neurosci 2000;3:708–15.

[81] Mu Y, Poo MM. Spike timing-dependent LTP/LTD mediates visual experience-dependent plasticity in a developing retinotectal system. Neuron2006;50:115–25.

[82] Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et al. Impairedinsulin and insulin-like growth factor expression and signaling mechanismsin Alzheimer’s disease – is this type 3 diabetes? J Alzheimers Dis2005;7:63–80.

[83] Moroo I, Yamada T, Makino H, Tooyama I, McGeer PL, McGeer EG, et al. Loss ofinsulin receptor immunoreactivity from the substantia nigra pars compactaneurons in Parkinson’s disease. Acta Neuropathol 1994;87:343–8.

[84] Gasparini L, Gouras GK, Wang R, Gross RS, Beal MF, Greengard P, et al.Stimulation of beta-amyloid precursor protein trafficking by insulin reducesintraneuronal beta-amyloid and requires mitogen-activated protein kinasesignaling. J Neurosci 2001;21:2561–70.

[85] Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda OJ, et al. Insulin-degrading enzyme as a downstream target of insulin receptor signalingcascade: implications for Alzheimer’s disease intervention. J Neurosci2004;24:11120–6.

[86] Farris W, Mansourian S, Chang Y, Lindsley L, Eckman EA, Frosch MP, et al.Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo.Proc Natl Acad Sci USA 2003;100:4162–7.

[87] Zhao WQ, De Felice FG, Fernandez S, Chen H, Lambert MP, Quon MJ, et al.Amyloid beta oligomers induce impairment of neuronal insulin receptors.FASEB J 2008;22:246–60.

[88] De Felice FG, Vieira MN, Bomfim TR, Decker H, Velasco PT, Lambert MP, et al.Protection of synapses against Alzheimer’s-linked toxins: insulin signalingprevents the pathogenic binding of Abeta oligomers. Proc Natl Acad Sci USA2009;106:1971–6.

[89] Lee HK, Kumar P, Fu Q, Rosen KM, Querfurth HW. The insulin/Akt signalingpathway is targeted by intracellular beta-amyloid. Mol Biol Cell2009;20:1533–44.

[90] Schubert M, Gautam D, Surjo D, Ueki K, Baudler S, Schubert D, et al. Role forneuronal insulin resistance in neurodegenerative diseases. Proc Natl Acad Sci2004;101(9):3100–5.

[91] Sánchez-Chávez G, Peña-Rangel MT, Riesgo-Escovar JR, Martínez-Martínez A,Salceda R. Insulin stimulated-glucose transporter Glut 4 is expressed in theretina. PLoS One 2012;7(12):529–59.

[92] Cheng CM, Reinhardt RR, Lee WH, Joncas G, Patel SC, Bondy CA. Insulin-likegrowth factor 1 regulates developing brain glucose metabolism. Proc NatlAcad Sci USA 2000;97:10236–41.

[93] Bingham EM, Hopkins D, Smith D, Pernet A, Hallett W, Reed L, et al. The roleof insulin in human brain glucose metabolism: an 18fluoro deoxyglucosepositron emission tomography study. Diabetes 2002;51:3384–90.

[94] Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetesmellitus and the risk of dementia: The Rotterdam Study. Neurology1999;53:1937–42.

[95] Tanaka M, Sawada M, Yoshida S, Hanaoka F, Marunouchi T. Insulin preventsapoptosis of external granular layer neurons in rat cerebellar slice culture.Neurosci Lett 1995;199:37–40.

[96] Herzog KH, Bailey K, Barde YA. Expression of the BDNF gene in the developingvisual system of the chick. Development 1994;120:1643–9.

[97] Yamamori T. Localization of cholinergic differentiation factor/leukemiainhibitory factor mRNA in the rat brain and peripheral tissues. Proc NatlAcad Sci 1991;88:7298–302.

[98] Ballotti R, Nielsen FC, Pringle N, Kowalski A, Richardson WD, Obberghen EV,et al. Insulin-like growth factor I in cultured rat astrocytes: expression of thegene, and receptor tyrosine kinase. EMBO J 1987;6:3633–9.

[99] Meyer-Franke A, Kaplan MR, Pfrieger FW, Barres BA. Characterization of thesignaling interactions that promote the survival and growth of developingretinal ganglion cells in culture. Neuron 1995;15:805–19.

[100] Mill JF, Chao MV, Ishii DN. Insulin, insulin-like growth factor II, and nervegrowth factor effects on tubulin mRNA levels and neurite formation. ProcNatl Acad Sci USA 1985;82:7126–30.

[101] Ito M. Insulin or bFGF and C2 ceramide increase newborn rat retinal ganglioncell survival rate. Biochem Biophys Res Commun 2003;7:564–71.

[102] Begum N, Robinson LJ, Draznin B, Heidenreich KA. Protein phosphatase-1and -2a activities in cultured fetal chick neurons: differential regulationby insulin and insulin-like growth factor-I. Endocrinology 1993;133:2085–9.

[103] Qian W, Shi J, Yin X, Iqbal K, Grundke-Iqbal I, Gong CX, et al. PP2A regulatestau phosphorylation directly and also indirectly via activating GSK-3beta. JAlzheimers Dis 2010;19(4):1221–9.

[104] Rodrigues M, Waldbillig RJ, Rajagopalan S, Hackett J, LeRoith D, Chader GJ.Retinal insulin receptors: localization using a polyclonal anti-insulin receptorantibody. Brain Res 1988;443:389–94.

[105] Heidenreich KA, Toledo SP, Brunton LL, Watson MJ, Daniel-Issakani S,Strulovici B. Insulin stimulates the activity of a novel protein kinase C, PKC-e in cultured fetal chick neurons. J Biol Chem 1990;265:15076–82.

[106] Ugolini G, Cremisi F, Maffei L. TrkA, TrkB and p75 mRNA expression isdevelopmentally regulated in the rat retina. Brain Res 1995;704:121–4.

[107] Hockenbery DM, Oltvai ZN, Yin XM, Milliman CL, Korsmeyer SJ. Bcl-2functions in an antioxidant pathway to prevent apoptosis. Cell1993;75:241–51.

[108] Chen DF, Schneider GE, Martinou JC, Tonegawa S. Bcl-2 promotesregeneration of severed axons in mammalian CNS. Nature 1997;385:434–9.

[109] Shimizu S, Eguchi Y, Kamiike W, Funahashi Y, Mignon A, Lacronique V, et al.Bcl-2 prevents apoptotic mitochondrial dysfunction by regulating protonflux. Proc Natl Acad Sci USA 1998;95:1455–9.

[110] Fischer AJ, Reh TA. Identification of a proliferating marginal zone of retinalprogenitors in the postnatal chicken. Dev Biol 2000;220:197–210.

[111] Prada C, Puga J, Pérez-Méndez L, López P, Ramírez G. Spatial andtemporal patterns of neurogenesis in the chick retina. Eur J Neurosci1991;3:559–69.

[112] Jasoni CL, Walker MB, Morrison MD, Reh TA. A chicken acheate-scutehomolog (CASH-1) is expressed in a temporally and spatially discrete mannerin the developing nervous system. Development 1994;120:769–83.

[113] Weston BC, Aliabadi Z, White GL. Glaucoma – review for the vigilant clinician.Clin Rev 2000;10:59–74.

[114] Goldberg I. Relationship between intraocular pressure and preservation ofvisual field in glaucoma. Surv Ophthalmol 2003;48:S3–7.

[115] Kass MA, Heuer DK, Higginbotham EJ, Johnson CA, Keltner JL, Miller JP, et al.The Ocular Hypertension Treatment Study: a randomized trial determinesthat topical ocular hypotensive medication delays or prevents the onset ofprimary open-angle glaucoma. Arch Ophthalmol 2002;120:701–13.

[116] Tielsch JM, Katz J, Quigley HA, Javitt JC, Sommer A. Diabetes, intraocularpressure, and primary open-angle glaucoma in the Baltimore Eye Survey.Ophthalmology 1995;102:48–53.

[117] Dielemans I, de Jong PT, Stolk R, Vingerling JR, Grobbee DE, Hofman A.Primary open-angle glaucoma, intraocular pressure, and diabetes mellitus inthe general elderly population. The Rotterdam Study. Ophthalmology1996;103:1271–5.

[118] Mori K, Ando F, Nomura H, Sato Y, Shimokata H. Relationship betweenintraocular pressure and obesity in Japan. Int J Epidemiol 2000;29:661–6.

[119] Oh SW, Lee S, Park, Kim DJ. Elevated intraocular pressure is associated withinsulin resistance and metabolic syndrome. Diabetes Metab Res Rev2005;21:434–40.

[120] Hepburn DA, Fisher BM, Thomson I, Barrie T, Frier BM. Autonomicmechanisms underlying intraocular pressure changes during insulin-induced hypoglycaemia in normal human subjects: effects ofpharmacological blockade. Clin Sci (Lond) 1991;80(4):333–8.

[121] Wu SY, Leske MC. Associations with intraocular pressure in the Barbados EyeStudy. The Barbados Eye Study Group. Arch Ophthalmol 1997;115:1572–6.

[122] Leske MC, Podgor MJ. Intraocular pressure, cardiovascular risk variables, andvisual field defects. Am J Epidemiol 1983;118:280–7.

[123] Reaven GM. Role of insulin resistance in human disease. Banting lecture1988. Diabetes 1998;37:1595–607.

[124] Szkudelski T. The mechanism of alloxan and streptozotocin action in B cells ofthe rat pancreas. Physiol Res 2001;50(6):537–46.

[125] Plaschke K, Hoyer S. Action of the diabetogenic drug streptozotocin onglycolytic and glycogenolytic metabolism in adult rat brain cortex andhippocampus. Int J Dev Neurosci 1993;11(4):477–83.

[126] Duelli R, Schröck H, Kuschinsky W, Hoyer S. Intracerebroventricular injectionof streptozotocin induces discrete local changes in cerebral glucoseutilization in rats. Int J Dev Neurosci 1994;12(8):737–43.

[127] Hoyer S, Lee SK, Löffler T, Schliebs R. Inhibition of the neuronal insulinreceptor. An in vivo model for sporadic Alzheimer disease? Ann NY Acad Sci2000;920:256–8.

Page 10: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

544 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

[128] Hoyer S, Lannert H. Inhibition of the neuronal insulin receptor causesAlzheimer-like disturbances in oxidative/energy brain metabolism and inbehavior in adult rats. Ann NY Acad Sci 1999;893:301–3.

[129] de la Monte SM, Wands JR. Alzheimer’s disease is type 3 diabetes-evidencereviewed. J Diabetes Sci Technol 2008;2(6):1101–13.

[130] de la Monte SM, Tong M, Lester-Coll N, Plater Jr M, Wands JR. Therapeuticrescue of neurodegeneration in experimental type 3 diabetes: relevance toAlzheimer’s disease. J Alzheimers Dis 2006;10(1):89–109.

[131] Lester-Coll N, Rivera EJ, Soscia SJ, Doiron K, Wands JR, de la Monte SM.Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadicAlzheimer’s disease. J Alzheimers Dis 2006;9(1):13–33.

[132] Su JH, Deng G, Cotman CW. Bax protein expression is increased inAlzheimer’s bracorrelations with DNA damage, Bcl-2 expression, and brainpathology. J Neuropathol Exp Neurol 1997;56(1):86–93.

[133] Grünblatt E, Salkovic-Petrisic M, Osmanovic J, Riederer P, Hoyer S. Braininsulin system dysfunction in streptozotocin intracerebroventricularlytreated rats generates hyperphosphorylated tau protein. J Neurochem2007;101(3):757–70.

[134] Salkovic-Petrisic M, Tribl F, Schmidt M, Hoyer S, Riederer P. Alzheimer-likechanges in protein kinase B and glycogen synthase kinase-3 in rat frontalcortex and hippocampus after damage to the insulin signalling pathway. JNeurochem 2006;96(4):1005–15.

[135] Hong M, Lee VM. Insulin and insulin-like growth factor-1 regulate tauphosphorylation in cultured human neurons. J Biol Chem1997;272(31):19547–53.

[136] Chen GJ, Xu J, Lahousse SA, Caggiano NL, de la Monte SM. Transient hypoxiacauses Alzheimer-type molecular and biochemical abnormalities in corticalneurons: potential strategies for neuroprotection. J Alzheimers Dis2003;5(3):209–28.

[137] Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et al. Impairedinsulin and insulin-like growth factor expression and signaling mechanismsin Alzheimer’s disease – is this type 3 diabetes? J Alzheimers Dis2005;7(1):63–80.

[138] Rivera EJ, Goldin A, Fulmer N, Tavares R, Wands JR, de la Monte SM. Insulinand insulin-like growth factor expression and function deteriorate withprogression of Alzheimer’s disease: link to brain reductions in acetylcholine. JAlzheimers Dis 2005;8(3):247–68.

[139] Ashery-Padan R, Gruss P. Pax6 lights-up the way for eye development. CurrOpin Cell Biol 2001;13:706–14.

[140] Kozmik Z. The role of Pax genes in eye evolution. Brain Res Bull2008;75:335–9.

[141] Glaser T, Walton DS, Maas RL. Genomic structure, evolutionary conservationand aniridia mutations in the human PAX6 gene. Nat Genet 1992;2:232–9.

[142] Hill RE, Favor J, Hogan BL, Ton CC, Saunders GF, Hanson IM, et al. Mouse smalleye results from mutations in a paired-like homeobox-containing gene.Nature 1991;354:522–5.

[143] Hogan BL, Hirst EM, Horsburgh G, Hetherington CM. Small eye (Sey): a mousemodel for the genetic analysis of craniofacial abnormalities. Development1988;103(Suppl):115–9.

[144] Glaser T, Jepeal L, Edwards JG, Young SR, Favor J, Maas RL. PAX6 gene dosageeffect in a family with congenital cataracts, aniridia, anophthalmia andcentral nervous system defects. Nat Genet 1994;7:463–71.

[145] Baumer N, Marquardt T, Stoykova A, Ashery-Padan R, Chowdhury K, Gruss P.Pax6 is required for establishing naso-temporal and dorsal characteristics ofthe optic vesicle. Development 2002;129:4535–45.

[146] Lakowski J, Majumder A, Lauderdale JD. Mechanisms controlling Pax6isoform expression in the retina have been conserved between teleosts andmammals. Dev Biol 2007;307:498–520.

[147] Hsieh YW, Yang XJ. Dynamic Pax6 expression during the neurogenic cellcycle influences proliferation and cell fate choices of retinal progenitors.Neural Dev 2009;4:32.

[148] Grindley JC, Davidson DR, Hill RE. The role of pax-6 in eye and nasaldevelopment. Development 1995;121:1433–42.

[149] Davis-Silberman N, Kalich T, Oron KV, Marquardt T, Kroeber M, Tamm ER,et al. Genetic dissection of Pax6 dosage requirements in the developingmouse eye. Hum Mol Genet 2005;14:2265–76.

[150] Alm A, Nilsson SF. Uveoscleral outflow – a review. Exp Eye Res2009;88:760–8.

[151] Leske MC, Heijl A, Hussein M, Bengtsson B, Hyman L, Komaroff E. Factors forglaucoma progression and the effect of treatment: the early manifestglaucoma trial. Arch Ophthalmol 2003;121:48–56.

[152] Howell GR, Libby RT, Jakobs TC, Smith RS, Phalan FC, Barter JW, et al. Axons ofretinal ganglion cells are insulted in the optic nerve early in DBA/2Jglaucoma. J Cell Biol 2007;179:1523–37.

[153] Walther C, Gruss P. Pax-6, a murine paired box gene, is expressed in thedeveloping CNS. Development 1991;113:1435–49.

[154] Koroma BM, Yang JM, Sundin OH. The Pax-6 homeobox gene is expressedthroughout the corneal and conjunctival epithelia. Invest Ophthalmol Vis Sci1997;38:108–20.

[155] Hogan BL, Horsburgh G, Cohen J, Hetherington CM, Fisher G, Lyon MF. Smalleyes (Sey): a homozygous lethal mutation on chromosome 2 which affectsthe differentiation of both lens and nasal placodes in the mouse. J EmbryolExp Morphol 1986;97:95–110.

[156] Hogan BL, Hirst EM, Horsburgh G, Hetherington CM. Small eye (Sey): a mousemodel for the genetic analysis of craniofacial abnormalities. Development1988;103:115–9.

[157] Ton CC, Hirvonen H, Miwa H, Weil MM, Monaghan P, Jordan T, et al.Positional cloning and characterization of a paired box- and homeobox-containing gene from the aniridia region. Cell 1991;67:1059–74.

[158] Prosser J, van Heyningen V. PAX6 mutations reviewed. Hum Mutat1998;11:93–108.

[159] Kumar K, Tanwar M, Naithani P, Insaan R, Garg S, Venkatesh P, et al. Pax6gene analysis in irido-fundal coloboma. Mol Vis 2011;17:1414–9.

[160] Walton DS. Unusual pediatric glaucomas. In: Epstein DL, editor. Chandler andgrant’s glaucoma. Baltimore, MD: Williams and Wilkins; 1997. p. 623–38.

[161] Nelson LB, Spaeth GL, Nowinski TS, Margo CE, Jackson L. Aniridia. A review.Surv Ophthalmol 1984;28:621–42.

[162] Gould DB, John SW. Anterior segment dysgenesis and the developmentalglaucomas are complex traits. Hum Mol Genet 2002;11:1185–93.

[163] Collins T. Congenital defects of the iris and glaucoma. Trans Ophthalmol SocUK 1893;13:128–39.

[164] Gosmain Y, Katz LS, Masson MH, Cheyssac C, Poisson C, Philippe J. Pax6 iscrucial for b-cell function, insulin biosynthesis, and glucose-induced insulinsecretion. Mol Endocrinol 2012;26(4):696–709.

[165] St Onge L, Sosa-Pineda B, Chowdhury K, Mansouri A, Gruss A. Pax6 is requiredfor differentiation of glucagon-producing alpha-cells in mouse pancreas.Nature 1997;387:406–9.

[166] Baulmann DC, Ohlmann A, Flügel-Koch C, Goswami S, Cvekl A, Tamm ER.Pax6 heterozygous eyes show defects in chamber angle differentiation thatare associated with a wide spectrum of other anterior eye segmentabnormalities. Mech Dev 2002;118(1–2):3–17.

[167] Margo CE. Congenital aniridia: a histopathologic study of the anteriorsegment in children. J Pediatr Ophthalmol Strabismus 1983;20:192–8.

[168] Gage PJ, Rhoades W, Prucka SK, Hjalt T. Fate maps of neural crest andmesoderm in the mammalian eye. Invest Ophthalmol Vis Sci2005;46:4200–8.

[169] Matsuo T, Osumi-Yamashita N, Noji S, Ohuchi H, Koyama E, Myokai F, et al. Amutation in the Pax-6 gene in rat small eye is associated with impairedmigration of midbrain crest cells. Nat Genet 1993;3:299–304.

[170] Collinson JM, Quinn JC, Buchanan MA, Kaufman MH, Wedden SE, West JD,et al. Primary defects in the lens underlie complex anterior segmentabnormalities of the Pax6 heterozygous eye. Proc Natl Acad Sci USA2001;98:9688–93.

[171] Jorgensen MC, Ahnfelt-Ronne J, Hald J, Madsen OD, Serup P, Hecksher-Sorensen J. An illustrated review of early pancreas development in themouse. Endocrine Rev 2007;28:685–705.

[172] Pan FC, Wright C. Pancreas organogenesis: from bud to plexus to gland. DevDyn 2011;240:530–65 [An Official Publication of the American Association ofAnatomists].

[173] Simpson TI, Price DJ. Pax6; a pleiotropic player in development. Bioessays2002;24:1041–51.

[174] St-Onge L, Sosa-Pineda B, Chowdhury K, Mansouri A, Gruss P. Pax6 is requiredfor differentiation of glucagon-producing alpha-cells in mouse pancreas.Nature 1997;387:406–9.

[175] Sander M, Neubüser A, Kalamaras J, Ee HC, Martin GR, German MS. Geneticanalysis reveals that PAX6 is required for normal transcription of pancreatichormone genes and islet development. Genes Dev 1997;11(13):1662–73.

[176] Ahlgren U, Jonsson J, Edlund H. The morphogenesis of the pancreaticmesenchyme is uncoupled from that of the pancreatic epithelium in IPF1/PDX1-deficient mice. Development 1996;122:1409–16.

[177] Gosmain Y, Marthinet E, Cheyssac C, Guerardel A, Mamin A, Katz LS, et al.Pax6 controls the expression of critical genes involved in pancreatic alphacell differentiation and function. J Biol Chem 2010;285:33381–93.

[178] Kaneko Y, Matsumoto G, Hanyu Y. Pax-6 expression during retinalregeneration in the adult newt. Dev Growth Differ 1999;41(6):723–9.

[179] Hamasaki A, Yamada Y, Kurose T, Ban N, Nagashima K, Takashashi A, et al.Adult pancreatic islets require differential pax6 gene dosage. BiochemBiophys Research Commun 2007;353:40–6.

[180] Ding J, Gao Y, Zhao J, Yan H, Guo SY, Zhang QX, et al. Pax6 haploinsufficiencycauses abnormal metabolic homeostasis by down-regulating glucagon-likepeptide 1 in mice. Endocrinology 2009;150:2136–44.

[181] Osumi N, Shinohara H, Numayama-Tsuruta K, Maekawa M. Concisereview: Pax6 transcription factor contributes to both embryonic andadult neurogenesis as a multifunctional regulator. Stem Cells 2008;26:1663–72.

[182] Wen JH, Chen YY, Song SJ, Ding J, Gao Y, Hu QK, et al. Paired box 6 (PAX6)regulates glucose metabolism via proinsulin processing mediated byprohormone convertase 1/3 (PC1/3). Diabetologia 2009;52:504–13.

[183] Hart AW, Mella S, Mendrychowski J, van Heyningen V, Kleinjan DA. Thedevelopmental regulator Pax6 is essential for maintenance of islet cellfunction in the adult mouse pancreas. PLoS ONE 2013;8(1):54173.

[184] Andersen FG, Jensen J, Heller RS, Petersen HV, Larsson LI, Madson OD, et al.Pax6 and Pdx1 form a functional complex on the rat somatostatin geneupstream enhancer. FEBS Lett 1999;445:315–20.

[185] Nitta Y, Shigeyoshi Y, Nakagata N, Kaneko T, Nitta K, Harada T, et al. Kineticsof blood glucose in mice carrying hemizygous Pax6. Exp Anim/Jpn Assoc LabAnim Sci 2009;58:105–12.

[186] Yukihiro F, Jeannie WC, David SK, Zhang T, Scott P, Anthony TC, et al.Combined expression of Pax6 and Pdx1 are required for GIP production.Diabetes 2007;56(1):372.

[187] Steiner DF. On the discovery of precursor processing. Methods Mol Biol2011;768:3–11.

Page 11: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546 545

[188] Hart AW, Baeza N, Apelqvist A, Edlund H. Attenuation of FGF signalling inmouse beta-cells leads to diabetes. Nature 2000;408:864–8.

[189] Wostyn P, Audenaert K, De Deyn PP. Alzheimer’s disease and glaucoma: isthere a causal relationship? Br J Ophthalmol 2009;93:1557–9.

[190] Tamura H, Kawakami H, Kanamoto T, Kato T, Yokoyama T, Sasaki K, et al.High frequency of open-angle glaucoma in Japanese patients withAlzheimer’s disease. J Neurol Sci 2006;246:79–83.

[191] McKinnon SJ. Glaucoma: ocular Alzheimer’s disease? Front Biosci2003;8:1140–56.

[192] Hoar RM. Embryology of the eye. Environ Health Perspect 1982;44:31–4.[193] Dickerman RD, Smith GH, Langham-Roof L, McConathy WJ, East JW, Smith

AB. Intra-ocular pressure changes during maximal isometric contraction:does this reflect intra-cranial pressure or retinal venous pressure? Neurol Res1999;21:243–6.

[194] Bol P. Glaucoma. Ned Tijdschr Tandheelkd 2003;110:298–9.[195] Fautsch MP, Johnson DH. Aqueous humor outflow: what do we know? Where

will it lead us? Invest Ophthalmol Vis Sci 2006;47:4181–7.[196] Guo L, Salt TE, Luong V, Wood N, Cheung W, Maass A, et al. Targeting amyloid-

beta in glaucoma treatment. Proc Natl Acad Sci USA 2007;104:13444–9.[197] Wostyn P. Can chronic increased intracranial pressure or exposure to

repetitive intermittent intracranial pressure elevations raise your risk forAlzheimer’s disease? Med Hypoth 2004;62:925–30.

[198] Wostyn P, Audenaert K, De Deyn PP. Alzheimer’s disease: cerebral glaucoma?Med Hypoth 2010;74:973–7.

[199] Engelborghs S, De Vreese K, Van de Casteele T, Vanderstichele H, EverbroeckB, Cras P, et al. Diagnostic performance of a CSF-biomarker panel in autopsy-confirmed dementia. Neurobiol Aging 2008;29:1143–59.

[200] Yoneda S, Hara H, Hirata A, Fukushima M, Inomata Y, Tanihara H. Vitreousfluid levels of beta-amyloid(1–42) and tau in patients with retinal diseases.Jpn J Ophthalmol 2005;49:106–8.

[201] McKinnon SJ, Lehman DM, Kerrigan-Baumrind LA, et al. Caspase activationand amyloid precursor protein cleavage in rat ocular hypertension. InvestOphthalmol Vis Sci 2002;43:1077–87.

[202] Liadis N, Murakami K, Eweida M, Elford AR, Sheu L, Gaisano HY, et al.Caspase-3-dependent b-cell apoptosis in the initiation of autoimmunediabetes mellitus. Mol Cell Biol 2005;25(9):3620–9.

[203] Wostyn P. Intracranial pressure and Alzheimer’s disease: a hypothesis. MedHypoth 1994;43:219–22.

[204] Wostyn P, Audenaert K, De Deyn PP. Alzheimer’s disease-related changes indiseases characterized by elevation of intracranial or intraocular pressure.Clin Neurol Neurosurg 2008;110:101–9.

[205] Pasquier F, Boulogne A, Leys D, Fontaine P. Diabetes mellitus and dementia.Diabetes Metab 2006;32(5 Pt 1):403–14.

[206] Nicolls MR. The clinical and biological relationship between type II diabetesmellitus and Alzheimer’s disease. Curr Alzheimer Res 2004;1(1):47–54.

[207] Myers MG, Sun XJ, White MF. The IRS-1 signaling system. Trends Biochem Sci1994;19(7):289–93.

[208] Ullrich A, Bell JR, Chen EY, Herrera R, Petruzzelli LM, Dull TJ, et al. Humaninsulin receptor and its relationship to the tyrosine kinase family ofoncogenes. Nature 1985;313(6005):756–61.

[209] Sun XJ, Crimmins DL, Myers Jr MJ, Miralpeix M, White MF. Pleiotropic insulinsignals are engaged by multisite phosphorylation of IRS-1. Mol Cell Biol1993;13(12):7418–28.

[210] Burgering BM, Coffer PJ. Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature 1995;376(6541):599–602.

[211] Kulik G, Klippel A, Weber MJ. Antiapoptotic signalling by the insulin-likegrowth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol1997;17(3):1595–606.

[212] Mill JF, Chao MV, Ishii DN. Insulin, insulin-like growth factor II, and nervegrowth factor effects on tubulin mRNA levels and neurite formation. ProcNatl Acad Sci USA 1985;82(20):7126–30.

[213] Puro DG, Agardh E. Insulin-mediated regulation of neuronal maturation.Science 1984;225(4667):1170–2.

[214] Craft S. Insulin resistance and Alzheimer’s disease pathogenesis: potentialmechanisms and implications for treatment. Curr Alzheimer Res2007;4(2):147–52.

[215] Haan MN. Therapy insight: type 2 diabetes mellitus and the risk of late-onsetAlzheimer’s disease. Nat Clin Pract Neurol 2006;2(3):159–66.

[216] Reger MA, Watson GS, Frey WH, Baker LD, Cholerton B, Keeling ML, et al.Effects of intranasal insulin on cognition in memory-impaired older adults:modulation by APOE genotype. Neurobiol Aging 2006;27(3):451–8.

[217] Winocur G, Greenwood CE. Studies of the effects of high fat diets on cognitivefunction in a rat model. Neurobiol Aging 2005;26(Suppl 1):46–9.

[218] Winocur G, Greenwood CE, Piroli GG, Grillo CA, Reznikov LR, Reagan LP, et al.Memory impairment in obese Zucker rats: an investigation of cognitivefunction in an animal model of insulin resistance and obesity. Behav Neurosci2005;119(5):1389–95.

[219] Hinton DR, Sadun AA, Blanks JC, Miller CA. Optic-nerve degeneration inAlzheimer’s disease. N Engl J Med 1986;315(8):485–7.

[220] Parisi V. Correlation between morphological and functional retinalimpairment in patients affected by ocular hypertension, glaucoma,demyelinating optic neuritis and Alzheimer’s disease. Semin Ophthalmol2003;18(2):50–7.

[221] Crish SD, Sappington RM, Inman DM, Horner PJ, Calkins DJ. Distal axonopathywith structural persistence in glaucomatous neurodegeneration. Proc NatlAcad Sci USA 2010;107(11):5196–201.

[222] Gupta N, Ly T, Zhang Q, Kaufman PL, Weinreb RN, Yucel YH. Chronic ocularhypertension induces dendrite pathology in the lateral geniculate nucleus ofthe brain. Exp Eye Res 2007;84(1):176–84.

[223] Gupta N, Ang LC, de Tilly LN, Bidaisee L, Yucel YH. Human glaucoma andneural degeneration in intracranial optic nerve, lateral geniculate nucleus,and visual cortex. Br J Ophthalmol 2006;90(6):674–8.

[224] Tsolaki F, Gogaki E, Tiganita S, Skatharoudi C, Lopatatzidi C, Topouzis F, et al.Alzheimer’s disease and primary open-angle glaucoma: is there aconnection? Clin Ophthalmol. 2011;5:887–90.

[225] Flammer J, Mozaffarieh M. What is the present pathogenetic concept ofglaucomatous optic neuropathy? Surv Ophthalmol 2007;52(Suppl2):162–73.

[226] Gupta N, Greenberg G, deTilly LN, Gray B, Polemidiotis M, Yucel YH. Atrophyof the lateral geniculate nucleus in human glaucoma detected by magneticresonance imaging. Br J Ophthalmol 2009;93:56–60.

[227] Yucel Y, Gupta N. Glaucoma of the brain: a disease model for the study oftranssynaptic neural degeneration. Prog Brain Res 2008;173:465–78.

[228] Heijl A, Leske MC, Bengtsson B, Hyman L, Hussein M. Reduction of intraocularpressure and glaucoma progression: results from the Early ManifestGlaucoma Trial. Arch Ophthalmol 2002;120:1268–79.

[229] Leske MC, Heijl A, Hussein M, Bengtsson B, Hyman L, Komaroff E. Factors forglaucoma progression and the effect of treatment: the early manifestglaucoma trial. Arch Ophthalmol 2003;121:48–56.

[230] Shields MB. Normal-tension glaucoma: is it different from primary open-angle glaucoma? Curr Opin Ophthalmol 2008;19:85–8.

[231] Bayer AU, Ferrari F, Erb C. High occurrence rate of glaucoma among patientswith Alzheimer’s disease. Eur Neurol 2002;47:165–8.

[232] Yochim BP, Mueller AE, Kane KD, Kahook MY. Prevalence of cognitiveimpairment, depression, and anxiety symptoms among older adults withglaucoma. J Glaucoma 2012;21:250–4.

[233] Kessing LV, Lopez AG, Andersen PK, Kessing SV. No increased risk ofdeveloping Alzheimer disease in patients with glaucoma. J Glaucoma2007;16:47–51.

[234] Sivak JM. The aging eye: Common degenerative mechanisms between theAlzheimer’s brain and retinal diseases. Invest Ophthalmol Vis Sci2013;54(1):871–80.

[235] Blanks JC, Hinton DR, Sadun AA, Miller CA. Retinal ganglion cell degenerationin Alzheimer’s disease. Brain Res 1989;6:364–72.

[236] Blanks JC, Torigoe Y, Hinton DR, Blanks RH. Retinal pathology in Alzheimer’sdisease. I. Ganglion cell loss in foveal/parafoveal retina. Neurobiol Aging1996;17:377–84.

[237] Hedges TR, Perez Galves R, Speigelman D, Barbas NR, Peli E, Yardley CJ.Retinal nerve fiber layer abnormalities in Alzheimer’s disease. ActaOphthalmol Scand 1996;74:271–5.

[238] Blanks JC, Schmidt SY, Torigoe Y, Porrello KV, Hinton DR, Blanks RH. Retinalpathology in Alzheimer’s disease. II. Regional neuron loss and glial changes inGCL. Neurobiol Aging 1996;17:385–95.

[239] Moore D, Harris A, Wudunn D, Kheradiya N, Siesky B. Dysfunctionalregulation of ocular blood flow: a risk factor for glaucoma? ClinOphthalmol 2008;2:849–61.

[240] Orgul S. Blood flow in glaucoma. Br J Ophthalmol 2007;91:3–5.[241] Berisha F, Feke GT, Trempe CL, McMeel JW, Schepens CL. Retinal

abnormalities in early Alzheimer’s disease. Invest Ophthalmol Vis Sci2007;48:2285–9.

[242] Tezel G, Wax MB. Hypoxia-inducible factor 1alpha in the glaucomatousretina and optic nerve head. Arch Ophthalmol 2004;122:1348–56.

[243] Gupta N, Fong J, Ang LC, Yucel YH. Retinal tau pathology in humanglaucomas. Can J Ophthalmol 2008;43:53–60.

[244] Loeffler KU, Edward DP, Tso MO. Tau-2 immunoreactivity of corporaamylacea in the human retina and optic nerve. Invest Ophthalmol Vis Sci1993;34:2600–3.

[245] Gupta N, Fong J, Ang LC, Yücel YH. Retinal tau pathology in humanglaucomas. Can J Ophthalmol 2008;43(1):53–60.

[246] Reddy PH, Manczak M, Mao P, Calkins MJ, Reddy AP, Shirendeb U. Amyloid-band mitochondria in aging and Alzheimer’s disease: implications forsynaptic damage and cognitive decline. J Alzheimers Dis 2010;20:499–512.

[247] Blanks JC, Schmidt SY, Torigoe Y, Porrello KV, Hinton DR, Blanks RH. Retinalpathology in Alzheimer’s disease. II. Regional neuron loss and glial changes inGCL. Neurobiol Aging 1996;17(3):385–95.

[248] Faiq MA, Saluja D, Qadri R. Encephalization – an evolutionary predispositionto diabetes: a ‘‘Large Brain Hypothesis’’ explaining the mechanism ofdiabetes. IOSR J Pharm Biol Sci 2013;5(6):66–72.

[249] Kirk CE. Visual influences on primate encephalization. J Hum Evol2006;51:76–90.

[250] Iwangoff P, Armbruster R, Enz A, Meier-Ruge W. Glycolytic enzymes fromhuman autoptic brain cortex: normal aged and demented cases. Mech AgeingDev 1980;14(1–2):203–9.

[251] Sims NR, Bowen DM, Smith CC, Flack RH, Davison AN, Snowden JS, et al.Glucose metabolism and acetylcholine synthesis in relation to neuronalactivity in Alzheimer’s disease. Lancet 1980;1(8164):333–6.

[252] Hoyer S. Causes and consequences of disturbances of cerebral glucosemetabolism in sporadic Alzheimer disease: therapeutic implications. Adv ExpMed Biol 2004;541:135–52.

[253] Schober MS, Chidlow G, Wood JP, Casson RJ. Bioenergetic-basedneuroprotection and glaucoma. Clin Exp Ophthalmol 2008;36(4):377–85.

Page 12: Glaucoma – Diabetes of the brain: A radical hypothesis about its nature and pathogenesis

546 M.A. Faiq et al. / Medical Hypotheses 82 (2014) 535–546

[254] Sperber GO, Bill A. Blood flow and glucose consumption in the optic nerve,retina and brain: effects of high intraocular pressure. Exp Eye Res1985;41(5):639–53.

[255] Virkamäki A, Ueki K, Kahn CR. Protein–protein interaction in insulin signalingand the molecular mechanisms of insulin resistance. J Clin Invest1999;103(7):931–43.

[256] Schubert M, Brazil DP, Burks DJ, Kushner JA, Ye J, Flint CL, et al. Insulinreceptor substrate-2 deficiency impairs brain growth and promotes tauphosphorylation. J Neurosci 2003;23(18):7084–92.

[257] Schubert M, Gautam D, Surjo D, Ueki K, Baudler S, Schubert D, et al. Role forneuronal insulin resistance in neurodegenerative diseases. Proc Natl Acad SciUSA 2004;101(9):3100–5.

[258] de la Monte SM, Ganju N, Banerjee K, Brown NV, Luong T, Wands JR. Partialrescue of ethanol-induced neuronal apoptosis by growth factor activation ofphosphoinositol-3-kinase. Alcohol Clin Exp Res 2000;24(5):716–26.

[259] de la Monte SM, Wands JR. Chronic gestational exposure to ethanol impairsinsulin-stimulated survival and mitochondrial function in cerebellar neurons.Cell Mol Life Sci 2002;59(5):882–93.

[260] Xu J, Yeon JE, Chang H, Tison G, Chen GJ, Wands J, et al. Ethanol impairsinsulin-stimulated neuronal survival in the developing brarole of PTENphosphatase. J Biol Chem 2003;278(29):26929–37.

[261] Ezzat VA, Duncan ER, Wheatcroft SB, Kearney MT. The role of IGF-1 and itsbinding proteins in the development of type 2 diabetes and cardiovasculardisease. Diabetes Obes Metab 2008;10(3):198–211.

[262] Calderari S, Gangnerau MN, Thibault M, Meile MJ, Kassis N, Alvarez C, et al.Defective IGF2 and IGF1R protein production in embryonic pancreas precedesbeta cell mass anomaly in the Goto-Kakizaki rat model of type 2 diabetes.Diabetologia 2007;50(7):1463–71.

[263] Liu JL. Does IGF-1 stimulate pancreatic islet cell growth? Cell BiochemBiophys 2007;48(2–3):115–25.

[264] Braak H, Braak E. Diagnostic criteria for neuropathologic assessment ofAlzheimer’s disease. Neurobiol Aging 1997;18(4 Suppl):S85–8.

[265] Nagy Z, Yilmazer-Hanke DM, Braak H, Braak E, Schultz C, Hanke J. Assessmentof the pathological stages of Alzheimer’s disease in thin paraffin sections: acomparative study. Dement Geriatr Cogn Disord 1998;9(3):140–4.

[266] de la Monte SM, Neely TR, Cannon J, Wands JR. Ethanol impairs insulin-stimulated mitochondrial function in cerebellar granule neurons. Cell MolLife Sci 2001;58(12–13):1950–60.

[267] de la Monte SM, Chen GJ, Rivera E, Wands JR. Neuronal thread proteinregulation and interaction with microtubule-associated proteins in SH-Sy5yneuronal cells. Cell Mol Life Sci 2003;60(12):2679–91.

[268] Soscia SJ, Tong M, Xu XJ, Cohen AC, Chu J, Wands JR, et al. Chronic gestationalexposure to ethanol causes insulin and IGF resistance and impairsacetylcholine homeostasis in the brain. Cell Mol Life Sci 2006;63(17):2039–56.

[269] de la Monte SM, Wands JR. Molecular indices of oxidative stress andmitochondrial dysfunction occur early and often progress with severity ofAlzheimer’s disease. J Alzheimers Dis 2006;9(2):167–81.

[270] Blasko I, Stampfer-Kountchev M, Robatscher P, Veerhuis R, Eikelenboom P,Grubeck-Loebenstein B. How chronic inflammation can affect the brain andsupport the development of Alzheimer’s disease in old age: the role ofmicroglia and astrocytes. Aging Cell 2004;3(4):169–76.

[271] Eikelenboom P, van Gool WA. Neuroinflammatory perspectives on the twofaces of Alzheimer’s disease. J Neural Transm 2004;111(3):281–94.

[272] Tuppo EE, Arias HR. The role of inflammation in Alzheimer’s disease. Int JBiochem Cell Biol 2005;37(2):289–305.

[273] Lorenzo A, Yankner BA. Amyloid fibril toxicity in Alzheimer’s disease anddiabetes. Ann NY Acad Sci 1996;777:89–95.

[274] Niikura T, Hashimoto Y, Tajima H, Nishimoto I. Death and survival of neuronalcells exposed to Alzheimer’s insults. J Neurosci Res 2002;70(3):380–91.

[275] Tsukamoto E, Hashimoto Y, Kanekura K, Niikura T, Aiso S, Nishimoto I.Characterization of the toxic mechanism triggered by Alzheimer’s amyloid-beta peptides via p75 neurotrophin receptor in neuronal hybrid cells. JNeurosci Res 2003;73(5):627–36.

[276] Yasuhara O, Tooyama I, Aimi Y, Bellier JP, Hisano T, Matsuo A, et al.Demonstration of cholinergic ganglion cells in rat retina: expression of an alternativesplice variant of choline acetyltransferase. J Neurosci 2003;23(7):2872–81.

[277] Jeon CJ, Strettoi E, Masland RH. The major cell populations of the mouseretina. J Neurosci 1998;18:8936–46.

[278] Hashimoto K. Can minocycline prevent the onset of Alzheimer’s disease? AnnNeurol 2011;69:739 [author reply 739–40].

[279] Parachikova A, Vasilevko V, Cribbs DH, LaFerla FM, Green KN. Reductions inamyloid-beta-derived neuroinflammation, with minocycline, restorecognition but do not significantly affect tau hyperphosphorylation. JAlzheimers Dis 2010;21:527–42.

[280] Song Y, Wei EQ, Zhang WP, Ge Q, Liu JR, Wang ML, et al. Minocycline protectsPC12 cells against NMDA-induced injury via inhibiting 5-lipoxygenaseactivation. Brain Res 2006;1085:57–67.

[281] Baptiste DC, Powell KJ, Jollimore CA, Hamilton C, LeVatte TL, Archibald ML,et al. Effects of minocycline and tetracycline on retinal ganglion cell survivalafter axotomy. Neuroscience 2005;134:575–82.

[282] Shimazawa M, Yamashima T, Agarwal N, Hara H. Neuroprotective effects ofminocycline against in vitro and in vivo retinal ganglion cell damage. BrainRes 2005;1053:185–94.

[283] Bosco A, Inman DM, Steele MR, Wu G, Soto I, Marsh-Armstrong N, et al.Reduced retina microglial activation and improved optic nerve integrity withminocycline treatment in the DBA/2J mouse model of glaucoma. InvestOphthalmol Vis Sci 2008;49:1437–46.

[284] Illum L. Transport of drugs from the nasal cavity to the central nervoussystem. Eur J Pharm Sci 2000;11(1):1–18.