Você está na página 1de 13

The Ocular Surface 16 (2018) 45e57

Contents lists available at ScienceDirect

The Ocular Surface


journal homepage: www.theocularsurface.com

Review

The impact of diabetes on corneal nerve morphology and ocular


surface integrity
Maria Markoulli a, *, Judith Flanagan a, b, Shyam Sunder Tummanapalli a, Jenny Wu a,
Mark Willcox a
a
School of Optometry & Vision Science, University of New South Wales, Sydney, Australia
b
Brien Holden Vision Institute, Sydney, Australia

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

Article history: Diabetes mellitus is a chronic disease that results from inadequate insulin production or ineffective in-
Received 18 January 2017 sulin utilization. It is one of the most common systemic diseases worldwide with increasing prevalence.
Received in revised form Diabetes mellitus is associated with premature mortality, macrovascular complications such as cardio-
3 October 2017
vascular disease, and microvascular complications, including nephropathy leading to kidney failure,
Accepted 26 October 2017
potentially blinding diabetic retinopathy, and diabetic neuropathy. While the retinal complications of
diabetes are well recognized by eye care professionals, the effects on the ocular surface are poorly un-
Keywords:
derstood. Recent studies have reported on the association between peripheral neuropathy and corneal
Corneal erosions
Corneal nerves
neuropathy, showing the latter to be of predictive value for the systemic disease. Corneal neuropathy can
Corneal neuropathy lead to loss of corneal sensation and can ultimately result in neurotrophic ulcers and significant visual
Diabetes morbidity. The epithelial fragility and poor wound healing that result from reduced epithelial adhesion to
In vivo confocal microscopy the underlying basement membrane in diabetes, together with corneal neuropathy, are thought to in-
Neuropathy crease the susceptibility to persistent corneal erosions and infection, as well as to increase the risk of
Neuropeptides post-surgical complications. The aim of this article is to review the impact of diabetes on corneal nerve
Ocular microbiome morphology and ocular surface integrity. Changes in the tear film and ocular surface microbiome are
Ocular surface
highlighted in discussion of the mechanisms that underpin ocular surface changes that increase the
Tears
susceptibility to corneal erosion and infection.
© 2017 Elsevier Inc. All rights reserved.

1. Introduction The WHO classifies diabetes into three main categories [5], with
other sub-categories also described in the literature. The first
1.1. Overview category, type 1, comprises 5e10% of the diabetic population, with
an age of onset usually before 30 years; the underlying cause is
The World Health Organization (WHO) defines diabetes mellitus destruction and loss of the secretory function of insulin-producing
as “a chronic disease that occurs when the pancreas does not pro- pancreatic b-cells. The genetic characterization of maturity-onset
duce enough insulin, or when the body cannot effectively use the diabetes in the young (MODY) reviewed by Tuomi et al. [6] fea-
insulin it produces.” [1] It is one of the most common systemic tures defective control of insulin secretion and introduces another
diseases in the world, with the American Diabetes Association type of insulin deficiency.
estimating that 25.8 million children and adults, approximately 8.3% The second category, type 2 diabetes, affects approximately 95%
of the population of the United States of America, have the condition of the diabetic population with increasing prevalence [3], described
[2]. Worldwide, diabetes is thought to affect 415 million people [3] by some as the pandemic of the 21st century [7]. This form of
and its prevalence continues to rise [4], with estimates that dia- diabetes is a result of inadequate insulin production or utilization
betes will affect more than 640 million people by the year 2040 [3]. [8]. Risk factors include family history, being overweight, older age,
ethnicity, gestational diabetes (shortage of insulin or pregnancy
hormones), impaired glucose tolerance or impaired fasting glucose,
* Corresponding author. School of Optometry & Vision Science, The University of hypertension, and abnormal cholesterol levels. In many instances,
New South Wales, Sydney, NSW, 2052, Australia. there is uncertainty regarding the diagnosis of type 2 diabetes, for
E-mail address: m.markoulli@unsw.edu.au (M. Markoulli).

https://doi.org/10.1016/j.jtos.2017.10.006
1542-0124/© 2017 Elsevier Inc. All rights reserved.
46 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

Abbreviations NADPH nicotinamide adenine dinucleotide phosphate


NF-kB nuclear factor-kB
AGE advanced glycation end products NGF nerve growth factor
AMP adenosine monophosphate NO nitric oxide
CGRP calcitonin gene-related peptide NPY neuropeptide Y
COX cyclooxygenase NT-3 neurotrophin-3
DAG diacylglycerol PKC protein kinase C
DPN diabetic peripheral neuropathy PMNs polymorphonuclear leukocytes
eNOs nitric oxide synthase PRK photorefractive keratectomy
Epi-LASIK epithelial laser in situ keratomileusis ROS reactive oxygen species
IGF-1 insulin-like growth factor SDS-PAGE sodium dodecyl sulfate polyacrylamide gel
IGFBP3 insulin-like growth factor binding protein 3 electrophoresis
IGF-1R insulin-like growth factor receptor TGF-b transforming growth factor-b
IL-1 interleukin 1 TIMP tissue inhibitors of metalloproteinase
LADA latent autoimmune diabetes in adults TNF-a tumor necrosis factor- a
LASEK laser epithelial keratomileusis VEGF vascular endothelial growth factor
LASIK laser in situ keratomileusis VIP vasoactive-intestinal peptide
MMPs matrix metalloproteinases WHO World Health Organization
MODY maturity-onset diabetes in the young ZO zonula occludens
Naþ/Kþ ATPase sodium potassium adenosine triphosphatase

example, in those who are not overweight and do not have meta- wound healing [31,32], increased susceptibility to persistent
bolic syndrome and in those with residual b-cell function [6]. This epithelial erosions [26], and hence propensity to corneal infection
confusion has led to the introduction of latent autoimmune dia- [33,34].
betes in adults (LADA) [9], thought to occur in 2e12% of all cases of
diabetes [10]. 1.2. Aim
The third diabetic category is known as pre-diabetes [11e13],
and is considered to be a substantial risk factor for progression to The aim of this article is to review the impact of diabetes on
type 2 diabetes and associated complications [14]. It is thought to corneal nerve morphology and ocular surface integrity. Changes in
affect between 10 and 15% of the population [11]. the tear film and ocular surface microbiome are highlighted in
Diabetes is associated with premature mortality and with sys- discussion of the mechanisms that underpin these ocular surface
temic complications such as cardiovascular disease, and micro- changes that increase the susceptibility to corneal erosion and
vascular complications including retinopathy, nephropathy and, infection.
neuropathy [8], leading to organ and tissue damage in approxi- A literature review was conducted through PubMed Central
mately one third to one half of people with diabetes [15]. These using the following keywords: diabetes AND peripheral neuropa-
complications can have a significant impact on both immediate and thy, diabetes AND corneal neuropathy, diabetes AND corneal ero-
long-term healthcare costs [16]. Chronic hyperglycemia is the core sions, diabetes AND wound healing, diabetes AND tear film,
causative mechanism for each of these complications as a result of diabetes AND ocular surgery, diabetes AND contact lens wear and
changed metabolism, which in turn results from advanced glyca- diabetes AND microbiome. Articles were included up to January
tion end (AGE) products, abnormalities in signaling cascades, and 2017.
elevated production in reactive oxygen species (ROS), as well as
abnormalities in the stimulation of hemodynamic regulation sys-
tems [17]. By controlling hyperglycemia, this cascade can also be 2. Corneal neuropathy
controlled and the complications delayed [18e20].
In the diabetic eye, damage to the retinal capillaries leads to Corneal nerves bundles, derived from the nasociliary branch of
progressive capillary occlusion, which in turn results in retinal the ophthalmic division of the trigeminal nerve, enter the
ischemia. Gradually, ischemic retinal tissue elaborates signals to
stimulate new vessel proliferation. New vessels are prone to
leakage of intravascular fluid and are also prone to hemorrhage,
increasing the risk of blindness from retinal detachment secondary
to a fibrovascular membrane, and macular edema [21].
While the retinal complications of diabetes are well-recognized
by eye care professionals, the impact on the ocular surface is poorly
understood, despite the fact that up to 70% of people with diabetes
suffer from diabetic keratopathy during the course of their disease
[22e25]. This lack of understanding may be due to the varied
presentation of ocular surface disease in diabetes, ranging from dry
eye disease [26], through to neurotrophic ulcers (Fig. 1) [27]. Recent
work has revealed reduced corneal nerve density to be progressive
with duration of diabetes [28]. This change in morphology results
in functional changes such as loss of corneal sensitivity [29,30] and Fig. 1. A neurotrophic ulcer as a result of diabetes (courtesy of the L.V. Prasad Eye
is thought to contribute to subsequent events resulting in poor Institute, Hyderabad, India).
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 47

peripheral cornea in a radial fashion parallel to the corneal surface causing conduction block and demyelination of axons [45,47e50].
in the stroma and converge in an area known as the inferior whorl Neurotrophic factors such as neurotrophin-3 (NT-3) and nerve
(Fig. 2) [35]. The large nerve bundles then divide, taking a 90- growth factor (NGF) are also reduced in concentration by mito-
degree turn to penetrate Bowman's layer to form the corneal sub- chondrial damage [51,52]. In the diabetic cornea, 8-
basal nerve plexus between Bowman's membrane and the basal hydroxydeoxyguanosine has been measured in the diabetic rat
epithelial cells [36]. The small nerve fibers terminate in free nerve cornea as a marker of oxidative stress and has been found to be
endings in the corneal epithelium [36]. Unlike nerves elsewhere in increased relative to control rat corneas [53]. This indicates that the
the body, it is possible to image the corneal nerves in vivo using apoptosis of corneal cells seen in diabetes may be related to
in vivo confocal microscopy due to the transparency of the cornea oxidative stress.
[37]. Such imaging offers useful insight into the systemic impact of
diabetes [28,38,39]. 2.1.2. AGE products
Chronic hyperglycemia induces several pathological pathways Protein glycation leading to the formation of AGE products plays
that result in nerve damage that leads to neural loss in peripheral an important role in the pathogenesis of diabetic peripheral neu-
nerves, including the corneal nerves [40]. The following section ropathy [54,55]. Glycation is the non-enzymatic covalent binding of
reviews the pathogenesis of diabetic neuropathy in order to better a protein or lipoprotein with a sugar molecule such as fructose or
understand the pathophysiology of corneal neuropathy. glucose [56]. These non-enzymatically glycated proteins slowly
form crosslinked protein products with altered structure and
2.1. Pathogenesis of diabetic neuropathy function known as AGE products [44]. Myelin proteins of the nerves
are glycated in diabetes, resulting in structural and functional
A number of pathological pathways have been identified, which damage of the peripheral nerves and axonal degeneration [55]. AGE
intersect and complement each other in the process of diabetic deposition has been reported in perineurial collagen and in the
nerve damage, with both metabolic and vascular factors acting axoplasm of nerve fibers and Schwan cells of human diabetic pe-
synergistically in all stages of the disease [41,42]. These mecha- ripheral nerves, suggesting that the enhanced glycation may play a
nisms and pathways are summarized in Fig. 3 and include oxidative role in the development of diabetic neuropathy [57,58]. In the
stress, AGE products, the polyol pathway and protein kinase C (PKC) cornea, the accumulation of AGE products may be responsible for
activation [43e45]. the observed increased autofluorescence [59]. A higher expression
of AGE products has been reported in the diabetic rat cornea,
2.1.1. Oxidative stress indicating that this may be implicated in the development of dia-
Oxidative stress refers to the state in which the production of betic keratopathy as a result of apoptosis [53].
ROS exceeds the ability of any surrounding antioxidants to reduce
ROS to non-toxic substances. Oxidative stress can lead to tissue 2.1.3. The polyol pathway
damage [46]. In diabetes, elevated levels of extracellular glucose Hyperglycemia causes increased levels of intracellular glucose
increase the oxidative metabolism of glucose in mitochondria [44]. in nerve cells, leading to saturation of the normal glycolytic
Excessive entry of glucose causes surplus transport of electrons, pathway. Excess glucose is therefore shunted into the polyol
generating highly reactive ROS known as superoxides [44]. These pathway, known also as the sorbitol-aldose reductase pathway
superoxides cause a dangerous imbalance in the mitochondrial [60]. The excess glucose is then converted to sorbitol and fructose
electron transport chain, accompanied by reduced scavenging ca- by the enzymes aldose reductase and sorbitol dehydrogenase. In
pacity of antioxidant enzymes [44]. The larger mitochondrial vol- the process of reducing high intracellular glucose to sorbitol, aldose
ume in nerve fibers renders axons more susceptible to reductase consumes and depletes the cofactor nicotinamide
mitochondrial hyperglycemic injury with a depletion of energy, adenine dinucleotide phosphate (NADPH) [61]. In addition, the
nerve cell membrane is relatively impermeable to sorbitol and
fructose, which tend to accumulate within the nerve cell, increasing
osmotic stress [62]. This excess accumulation of sorbitol and fruc-
tose leads to reduced levels of free nerve myoinositol, a sugar
alcohol that is required for the normal nerve function [63]. A
reduction in myoinositol decreases membrane sodium potassium
adenosine triphosphatase (Naþ/Kþ ATPase) activity, which slows
down nerve conduction velocity and brings about nerve structural
breakdown. The polyol pathway is known to play a role in diabetic
cataract formation [64], but its role in corneal neuropathy is not yet
understood.

2.1.4. Protein kinase C activation pathway


Elevated intracellular glucose stimulates the synthesis of an
endogenous protein kinase C (PKC) activator, diacylglycerol (DAG)
[65,66]. Activation of PKC can affect Naþ/Kþ ATPase activity, and
other enzymes that are crucial in cellular membrane potential,
nerve conduction, and nerve regeneration [67e69].

2.2. Diabetic peripheral neuropathy

Diabetic neuropathy is characterized by progressive loss of pe-


ripheral nerve axons in a distal to proximal pattern, resulting in
pain, decreased sensation, and eventually complete loss of sensa-
Fig. 2. In vivo confocal microscopy image of the inferior corneal whorl. tion [70]. A simple classification based on anatomical and
48 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

Fig. 3. Pathophysiology of diabetic neuropathy: the underlying mechanisms of nerve damage as a result of hyperglycemia.

pathophysiological characteristics originally proposed by Thomas estimate [36]. In vivo confocal microscopy illuminates and images a
[71] and modified by Boulton [72] and Vinik [73,74] classifies dia- single point of tissue (400  400 mm) simultaneously in the same
betic neuropathies as peripheral, autonomic, proximal and focal. plane. The parameters measured include: nerve fiber bundle den-
Diabetic peripheral neuropathy is the most common neuropathic sity (number of nerve fiber bundles per mm2), nerve branch density
presentation in diabetes, comprising 75% of all diabetic neuropa- (number of nerve fiber branches per mm2), total nerve fiber length
thies [75], with painful diabetic peripheral neuropathy being pre- (per mm2), number of nerve interconnections (per mm2), nerve
sent in approximately 26% of patients with diabetes [76]. Following fiber width and nerve fiber tortuosity [37]. In healthy people
a 25-year follow-up, approximately 50% of patients with diabetes without diabetes, these variables have been found to be stable over
were found to have diabetic peripheral neuropathy [77]. Foot ul- a three-year period [90], except in contact lens wearers, in whom
ceration occurs in 7% of patients with diabetic peripheral neurop- reduced nerve density compared to non-lens wearers has been
athy compared with 1% of individuals with diabetes but without reported [91]. Age has also been shown to reduce corneal nerve
neuropathy [33]. Lower limb amputation is required in advanced fiber density [90,92,93]. A recent review by Cruzat et al. highlighted
cases [33,72,76,78]. There is currently no U.S. Food and Drug the corneal diseases that can affect corneal nerve parameters [94].
Administration (FDA)-approved therapy that prevents or reverses In diabetes, a reduction in corneal nerve fiber length, corneal nerve
diabetic peripheral neuropathy. Attention is therefore targeted at fiber density, and nerve fiber branch density has been found with
detection in order to identify those at increased risk of developing increasing neuropathic severity in both type 1 and type 2 diabetes
sequelae. (Fig. 4) [89,95]. Edwards and colleagues [83] showed that study
Diabetic peripheral neuropathy affects both the small and large participants with diabetic peripheral neuropathy had significantly
peripheral nerve fibers. The current gold standard for diagnosis of reduced corneal nerve fiber length and branch density compared to
diabetic peripheral neuropathy is vibration perception using bio- controls and participants with diabetes but without diabetic pe-
thesiometry [79]. However, biothesiometry measures large nerve ripheral neuropathy [83]. Pritchard et al. reported in a longitudinal
fiber function rather than small nerve fiber function, with changes study that reduction in corneal nerve fiber length is predictive of
to the latter being more prevalent in type 1 diabetes [80] and being the development of diabetic peripheral neuropathy [96], while
an earlier indicator of peripheral neuropathy [81]. Objective and Misra and colleagues reported that subbasal nerve density changes
sensitive assessment of small nerve fiber damage is via skin punch precede other clinical and electrophysiology tests of neuropathy
biopsy to examine nerve morphology [82]. Biopsy, however, is [39]. These findings are supported by a meta-analysis that included
invasive and non-repeatable [83], and the procedure can place 13 studies and 1680 participants and found that corneal nerve fiber
patients at risk of poor wound healing and infection [84]. The lack density, nerve branch density and nerve fiber length were signifi-
of a sensitive, non-invasive, and repeatable endpoint to measure cantly reduced in people with diabetic peripheral neuropathy [97].
changes in peripheral nerves is a major factor limiting clinical trials In the animal model, a study on the corneal dendritic cells in
for the treatment of diabetic peripheral neuropathy [33,85,86]. streptozotocin-diabetic mice identified greater corneal infiltration
Morphological changes in the corneal subbasal nerve plexus with dendritic cells compared to controls, as well as a significant
correlate with changes in the peripheral nerves and hence might negative correlation between the number of dendritic cells and
provide an alternate measure for diabetic peripheral neuropathy corneal nerve fiber density [98]. The close proximity of dendritic
[33,87e89]. cells with the subbasal nerve plexus and their function to recog-
nize, process, and present antigens indicates that the dendritic cells
2.3. Corneal neuropathy as an alternate measure of peripheral contribute to the development of corneal neuropathy [98].
neuropathy While diabetic neuropathy might be associated with reduced
nerve migration [99], that is, the movement of nerves relative to the
The corneal subbasal nerve plexus is the main object for study inferior whorl, recent studies have demonstrated that corneal
when assessing corneal nerve density using in vivo confocal mi- nerves in people with diabetes can regenerate after interventions to
croscopy. It is arranged in a single plane parallel to the corneal improve diabetic control [40,88,100]. Significant improvements in
surface, whereas stromal nerves are distributed throughout the nerve fiber length were seen 12 months after simultaneous
stroma in three dimensions, making their density difficult to pancreas and kidney transplantation in a group of 15 patients with
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 49

Fig. 4. A reduction in corneal nerve fiber density has been reported in patients with diabetes relative to healthy controls.

type 1 diabetes [78], and significant improvements in intra- free tears, or soft contact lens bandages [103]. More recent thera-
epidermal nerve fiber density were seen after 24 months of strict pies have been reviewed by Shih et al. [104] and offer promising
glycemic control and changes in lifestyle [101]. These studies sug- alternatives to protect against the loss of corneal sensitivity and the
gest that corneal nerve assessment may be a suitable index to subsequent development of corneal ulcers.
evaluate interventional efficacy. To understand the cascade of events that leads to the manifes-
While diabetic corneal neuropathy has significant implications tation of neurotrophic keratopathy, we review in the sections
for the diagnosis of diabetic peripheral neuropathy, it also has below how diabetes affects the corneal epithelium and the tear
significant local impact on the ocular surface itself. The following film. Given that dry eye disease predisposes the ocular surface to
section reviews the downstream effect of diabetic corneal neu- greater surface cell desquamation [105], the co-existence of dia-
ropathy on the integrity of the ocular surface and how neuropathy betes and dry eye disease places individuals at risk of thinning of
increases the susceptibility to injury and infection. the corneal epithelium [38]. When combined with a poor tear film,
the risk of epithelial damage is increased, disrupting the normal
3. Ocular surface integrity in diabetes barrier function of the epithelium [106]. Ocular surgery and contact
lens wear provide an additional challenge to an already compro-
Corneal nerves provide trophic support to the corneal epithelial mised ocular surface. The following sections also review the impact
cells by releasing soluble mediators that stimulate epithelial cell of these challenges on diabetic ocular surface integrity.
growth, mitosis, differentiation, and migration [36]. Epithelial cells,
in turn, provide trophic support to corneal neurons by secreting 3.1. Corneal epithelial wound healing in diabetes
growth factors that promote neurite extension [36]. In diabetes,
there is a reduction in the concentration of these mediators [38], Epithelial cells are pleomorphic and irregularly arranged in
leading to a disruption in epithelial integrity, and a flow-on effect diabetic eyes, with reduced epithelial cell proliferation and inade-
that causes further neuron loss, epithelial fragility, thinning of the quate adhesion of cells to an abnormal basement membrane [26].
epithelium and reduced epithelial cell density [38]. This vicious These irregularities translate to a cornea that is prone to persistent
cycle leads to the formation of recurrent corneal erosions and epithelial erosions with a weakened epithelial barrier [106,107].
eventually results in diabetic neurotrophic keratopathy [30]. When corneal neurons are damaged in diabetes, epithelial cells
Neurotrophic keratopathy is a severe manifestation of ocular swell, lose microvilli, and produce abnormal basal lamina [108,109].
surface dysfunction secondary to corneal denervation with an Epithelial cells slough off into tears at an accelerated rate and
estimated prevalence of 5/10,000 individuals [27]. Corneal nerve corneal staining appears within hours of denervation [36,110].
damage leads to neurotrophic keratopathy both directly (through Delayed wound healing has been shown in the streptozotocin rat
loss of trophic support) and indirectly (through dry eye from model in diabetes [111]. A 5-mm wound in the center of the cornea
reduction of tear and mucin production) [102]. Neurotrophic ker- healed in 100% of the healthy rats within 48 h, while only one-third
atopathy is classified into three stages. Stage 1 is characterized by of the diabetic rats healed within the same time [111]. Furthermore,
rose bengal staining of the inferior palpebral conjunctiva, a a diminished activation of endothelial growth factor receptor was
reduction in tear break-up time, and superficial corneal staining. In found in the rat diabetic corneas compared to healthy controls as
longstanding cases, superficial vascularization, stromal scarring, well as a different distribution in the expression of the tight junc-
and epithelial hyperplasia can follow. In stage 2, epithelial break- tion proteins [111]. The rat diabetic corneas had sparser b-catenin in
down occurs, with an epithelial defect surrounded by loose the apical cornea and minimal zonula occludens-1 (ZO-1) in the
epithelium that is hazy and edematous. Stage 2 presents with a central cornea [111]. The authors concluded that there was delayed
characteristic smooth and rolled appearance as the defect ages but not absent formation of cell-cell junctions in diabetic corneas.
without normal epithelial growth. Stage 3 is characterized by These epithelial changes in diabetic corneas are thought to be due
stromal involvement, with potential for stromal melting and to the direct effects of hyperglycemia on the cellular metabolism,
perforation [36]. Persistent epithelial defects are often unrespon- the accumulation of AGE products on the basement membrane
sive to conventional treatment, including patching, preservative- [112] and oxidative stress [36,113,114].
50 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

3.2. Tear film biochemistry in diabetes nerves are not related to the keratopathy seen in diabetes [134].
This study, however, measured the neuropeptides only 3 months'
The tear film is critical to the protection, cellular regeneration, after disease onset.
and wound healing of the ocular surface. These mediators are Topical treatment with derivatives of substance P and IGF-1
released by both the corneal nerves and the epithelial cells as have been shown to help heal epithelial defects and restore
trophic support. ocular surface integrity in neurotrophic keratopathies, including in
In diabetes, it is thought that there is a change in the distribution diabetic keratopathy [23,102]. In addition, topical NGF can restore
of these mediators, leading to a disruption in epithelial integrity, corneal sensitivity and ocular surface integrity after corneal neu-
and a flow on effect that causes further neuron loss and ultimately rotrophic keratitis [135,136]. NGF is a neurotrophin and is essential
neurotrophic ulcers [30,115,116]. An understanding of these un- for the survival and growth of sympathetic and sensory neurons
derlying mechanisms may contribute to the understanding ocular and for differentiation of neurons in the central nervous system.
surface disease in diabetes, as well as to the development of NGF also restores the function of damaged neurons and stimulated
treatments and preventative measures. The following sections re- mucin production by goblet cells [137]. There are receptors for NGF
view the biochemistry of the tear film and its influence on the on corneal and conjunctival epithelial cells [136].
ocular surface in diabetes.
3.2.2. Insulin
3.2.1. Neuropeptides and nerve growth factor Insulin was identified in the tear film, and insulin receptors were
Neuropeptides, including substance P, insulinelike growth found on the human ocular surface by Rocha and colleagues in
factor 1 (IGF-1), calcitonin gene-related peptide (CGRP), neuro- 2002 [138]. More recently, evidence was found for extra pancreatic
peptide Y (NPY) and vasoactive-intestinal peptide (VIP) [117], are insulin production by the lacrimal gland [139]. Whether the insulin
released from sensory nerve endings in the cornea, conjunctiva and found in tears is a product solely of the lacrimal gland or whether it
lacrimal gland [118]. Substance P and CGRP are the most abundant is pancreatic in origin is unknown [139]. Insulin supports the
neuropeptides in trigeminal ganglion neurons [119]. Neuropeptides metabolism and growth of exocrine glands, such as the lacrimal
can modulate the infiltration and activation of immune cells and gland, and plays a role in maintaining corneal and lacrimal
interact with endothelial, immune and mast cells [120] to play an epithelial cells, as well as meibomian gland cells [140], possibly
inflammatory role by inducing blood vessel dilation, leukocyte exerting a mitogenic stimulus in a similar manner to other growth
extravasation, immune cell activation, and synthesis and release of factors on the ocular surface [138,141,142]. Specific mechanisms of
several cytokines [117,121]. In the process, neuropeptides induce insulin action on the ocular surface, and how this action relates to
neurogenic inflammation symptoms, which manifest as dry eye glucose in tears is unclear [138]. In diabetes, a reduction in insulin-
disease [120]. In isolation and in combination with other constit- derived neurotrophic support has been proposed to contribute to
uents, substance P plays a role in the maintenance and nutrition of the corneal denervation observed in diabetic keratopathy [143].
the cornea by promoting migration and proliferation of corneal Several animal studies have reported that topical insulin promotes
epithelial cells [122e126]. Neuropeptides VIP and NPY can be anti- re-epithelialization and improved corneal sensitivity in diabetic
inflammatory, inhibiting T-cell proliferation and helper T-cell type rats with keratopathy [144e146]. Insulin-like growth factor binding
1 response while moderating the release of cytokines, chemokines protein 3 (IGFBP3) has been found to be upregulated 3-fold in type
and NO [117]. 2 diabetes, leading to insulin-like growth factor receptor (IGF-1R)
Substance P levels in the central and peripheral nervous system activation which, in turn, may lead to disruption of cellular adhe-
have been found to be significantly decreased in type 1 diabetes, sion, contributing to the pathogenesis of diabetic corneal compli-
particularly in patients with diabetic peripheral neuropathy [127]. cations [147].
In the tear film, the concentration of substance P has been corre-
lated with the duration of diabetes and severity of diabetic reti- 3.2.3. Glucose
nopathy [118], as well as corneal nerve fiber density [128,129]. In An increased concentration of glucose in the tears of patients
support of this, eyes with corneal hypoesthesia have also been re- with diabetes has been reported, with the tear glucose level
ported to have decreased concentrations of substance P compared correlating with the blood glucose level [148,149]. This correlation
to healthy control eyes [130]. Of note, adding non-steroidal anti- has led researchers to develop ocular surface glucose sensing de-
inflammatory drops such as diclofenac can further reduce the vices for non-invasive monitoring of blood glucose levels [150]. It is
concentration of substance P in tears [131]. This has implications for not currently known whether the increased glucose concentration
patients with corneal neuropathy requiring concurrent diclofenac in tears plays a role in hyperglycemic damage to corneal nerves in
or other non-steroidal anti-inflammatory drops. Diclofenac can patients with diabetes.
inhibit the cyclooxygenase (COX)-2 pathway of arachidonic acid
metabolism, ultimately blocking prostaglandin release [132]. With 3.2.4. AGE products
a reduction in substance P, corneal epithelial migration can Elevated levels of glucose in tears of individuals with diabetes
decrease and neurotrophic keratopathy may develop, resulting in [148,149] might result in the glycation of tear proteins, resulting in
further corneal complications in the case of people with diabetes AGE products. In a study of the basal tears of individuals with
[118]. In one published case study, a patient with diabetes was diabetes and controls by Zhao et al., total AGE-modified proteins
treated with collagen cross-linking for keratoconus and then were elevated in those with diabetes, both with and without reti-
treated with non-steroidal anti-inflammatory drops (nepafenac), nopathy [151]. The concentration of the tear film AGE modified
and presented 6 weeks post-operatively with corneal melting, proteins correlated with the AGE modified hemoglobin and the
perforation, and iris prolapse [133]. post-prandial blood glucose levels [151]. Western blots of the two-
In contrast to these reports on substance P, a study using the dimensional gels indicated that many protein species were AGE
diabetic mouse model found that while the concentrations of modified, more so in the diabetes samples. Zhao and colleagues
neuropeptides CGRP and VIP were elevated in the irides, there was suggest that these results indicate that tear film AGE-modified
no change in these levels in the corneas of the diabetic mice [134]. proteins may be used as a diagnostic biomarker for diabetes
The authors argue that the absence of change in these corneal [151]. It is not known, however, if glycation of tears impacts on tear
neuropeptides suggests that trophic peptides in corneal sensory film function or corneal nerves.
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 51

3.2.5. Abundant proteins due to reduction in mucin production by goblet cells. Reductions in
Grus and colleagues collected the tears from 255 controls and corneal innervation result in reduced goblet cell function [30].
260 people with diabetes but without anterior eye complications Dogru et al. and Inoue et al. have confirmed that diabetic patients
and analyzed the samples using sodium dodecyl sulfate poly- have decreased basal tear production and lacrimation, with the
acrylamide gel electrophoresis (SDS-PAGE), hence investigating the cotton thread test and Schirmer test indicating lacrimal gland
more abundant proteins [152]. The authors reported significantly involvement [30,168]. Dogru et al. further demonstrated that poor
more ‘peaks’ in people with diabetes compared to controls, and diabetic control and peripheral neuropathy caused significantly
more so in those with longer disease duration, indicating more lower tear lacrimation, postulating the theory that peripheral
proteins of higher concentration. This is in accordance with a study neuropathy affects the lacrimal gland [30]. A higher osmolarity has
by Stolwijk et al., who compared the tears of patients with and also been reported in patients with diabetes compared to controls
without diabetic retinopathy [153]. While total protein content did [169,170], although the reverse was found in a study by Beckman
not differ compared to healthy controls, lysozyme was significantly et al. [171] This is thought to be due to a reduction in lacrimal gland
greater in those with retinopathy and pre-albumin was found to be secretion or increased tear evaporation.
present in lower concentrations [153]. Immortalized human meibomian gland epithelial cells can be
stimulated by insulin in a dose-dependent manner to proliferate
3.2.6. Inflammatory mediators and accumulate lipid, while excess glucose causes progressive cell
Tumor necrosis factor-a (TNF-a) is an inflammatory cytokine loss and alterations to cell morphology [140]. These findings sup-
associated with insulin resistance [154]. TNF-a is a potent mediator port the hypothesis that insulin deficiency and hyperglycemia are
of leukostasis and is involved in the apoptotic pathway of retinal toxic for the meibomian gland epithelial cells, placing individuals
neurons and vascular endothelial cells in diabetic retinopathy, with diabetes at risk of meibomian gland dysfunction and dry eye
contributing to the breakdown in the blood-retinal-barrier. Cos- disease [140].
tagliola et al. measured the concentration of TNF-a in the tears of
people with and without type 2 diabetes using the Schirmer strip. 3.4. Ocular surface integrity after ocular surgery in diabetes
Participants with proliferative diabetic retinopathy had signifi-
cantly greater levels of TNF-a in their tears compared to those with 3.4.1. Cataract surgery
non-proliferative diabetic retinopathy, and healthy controls [155]. A common cause of visual impairment in diabetes is the for-
These authors suggest that TNF-a plays a role in the pathophysi- mation of a cataract, with a three-to four-fold increase in preva-
ology of diabetic retinopathy and suggest that TNF-a may be used lence of cataract in people with diabetes under the age of 65
as a means by which to screen for diabetic retinopathy [155]. [172,173]. As a result, phacoemulsification is commonly per-
Matrix metalloproteinases (MMPs) are a family of zinc- formed in patients with diabetes. Besides the well-known risks of
dependent endopeptidases found in tissues and fluids throughout post-operative progression of diabetic retinopathy and the devel-
the body. MMP-9 has an affinity for collagen VII, which is present in opment of macular edema, as reviewed by Haddad et al. [174],
the anchoring fibrils within the corneal basement membrane and is people with diabetes also have a greater risk of ocular surface
vital for corneal regeneration and wound healing [156]. Regulation complications and have an increased 30-day post-procedure
of MMPs involves endogenous tissue inhibitors of metal- readmission following cataract surgery compared to people
loproteinase (TIMPs) [156]. The ratio of MMP-9 to TIMP-1 is without diabetes [175]. Complications include superficial corneal
important in maintaining homeostasis [157]. Excess MMPs in tissue staining [22], corneal erosions [176], delayed wound healing [22]
or tears lead to ulceration and perforation in extreme cases [158]. and corneal edema [177], implying damage to the endothelium
Elevated MMP-9 levels in tissue have been shown to occur in [178,179]. Diabetes as a risk factor for endophthalmitis following
recurrent corneal erosions [159], dermatological ulcers and in cataract surgery has been both supported [180,181] and refuted
diabetic retinopathy [22,160]. [182,183].
Subbasal nerve density has also been found to be reduced
3.3. Diabetes and dry eye disease following cataract surgery [184]. Given the already reduced nerve
density in diabetes, surgery may further compromise the cornea,
In a healthy eye, superficial damage to the ocular surface is particularly when combined with reduced tear secretion [165] and
rapidly healed, with contributions to the healing process coming an abnormal basement membrane [22]. Jiang et al. reported that
from tissue and the tear film. In aqueous deficient dry eye, the individuals with diabetes undergoing phacoemulsification are
amount of tears is reduced which may increase the risk of corneal more prone to developing dry eye disease than their non-diabetic
erosion formation [36]. counterparts [185]. They found that 7 days post-operatively, 17%
Approximately 50% of people with diabetes complain of dry eye of the patients with diabetes developed dry eye disease, as indi-
symptoms [161e163]. With longer disease duration, reduced cated by tear film break-up time, Schirmer scores, and corneal
corneal sensation results in greater tolerance to dry eye disease, staining, compared to only 8% of the control patients [185]. While
reflecting the progression of the neuropathy [141]. Interestingly, the control group had a 0% incidence after 1 month post-
dry eye signs and symptoms have also been found to correlate with operatively, the group with diabetes took 3 months to return to
the degree of peripheral neuropathy and severity of diabetic reti- baseline levels [185]. These findings are supported by a retrospec-
nopathy [26, 162,164]. tive clinical study of the prevalence of dry eye in dogs post-cataract
There is evidence that tear film parameters are altered in pa- surgery, where the risk was greatest 2-weeks post-operatively in
tients with diabetes, with a reduction in tear break-up time and tear the case of diabetes [186].
secretion [165]. Factors thought to contribute to reduced tear pro- These findings indicate that the post-operative care of patients
duction in diabetes include microvascular damage to the lacrimal with diabetes should be customized and care given during surgery
gland from hyperglycemia, reduced binding sites for androgens and to minimize damage to the epithelium. Given the role of substance
estrogens on the lacrimal gland [166], reduced lacrimal innervation P and IGF-1 in promoting epithelial migration [121], Chikamoto and
due to autonomic neuropathy, lacrimal gland insufficiency [167], colleagues investigated the use of drops containing these sub-
and reduced reflex tearing due to impairment of corneal sensitivity stances on the prevention of corneal staining following cataract
[162]. Reduction in tear film stability and tear break-up time may be surgery [187]. When compared to saline eye drops, the substance P
52 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

e IGF-1 combination resulted in significantly less staining 2 days reports that diabetes is a risk factor for corneal complications
and 7 days following cataract surgery, with no difference found by following vitrectomy [197,198] and laser photocoagulation. Dogru
the two-week point [187]. As corneal staining represents the loss of and colleagues reported on the corneal response to argon green
the epithelial barrier, quick recovery is important for the preven- focal/grid laser photocoagulation using an applanation contact lens
tion of infection. Autologous serum has been used with similar and coupling fluid in a group of patients with diabetes and a group
effect [188]. Similarly, in a randomized, double-blind study without diabetes [199]. The group with diabetes had lower corneal
comparing the effect of oral aldose reductase inhibitor on post- sensitivity, lower tear film break-up time, and a greater extent of
surgical corneal staining in diabetes, an improvement in corneal corneal staining following the procedure, a finding supported by
staining, corneal sensitivity and symptoms was noted compared to Ozdemir et al. [176] Dogru et al. attribute this to the use of the
placebo, while measures of the tear film did not show a difference coupling fluid in conjunction with the co-existence of dry eye dis-
[189]. Topical aldose reductase has similarly been shown to ease [199]. The potential of topical anesthetic to prevent cell
improve corneal epithelial barrier function [190]. Given the role migration [200] may further add to this result.
that aldose reductase plays in the activation of the polyol pathway, These findings indicate that regardless of the form of ocular
its inhibition may reduce the damage to the corneal nerves and surgery, greater vigilance for corneal complications may be
hence allow normal wound healing to take place. The findings of required post-operatively when a patient presents with a history of
these studies are promising and may help to reduce the ocular diabetes.
surface complications seen after routine surgery.
3.5. Complications with contact lens use in diabetes
3.4.2. Refractive surgery
Refractive surgery offers a model for assessing the impact of Consensus is lacking among practitioners regarding the safety of
interfering with the corneal subbasal nerve plexus, via incision or contact lenses for patients with diabetes, with some considering
obliteration. In laser in situ keratomileusis (LASIK), a hinged flap of diabetes as a contraindication to lens wear [201]. When the actual
corneal epithelium and stroma is created with a keratome. Once the prescribing habits were surveyed in the United Kingdom, 2% of
flap is folded back, a laser is applied to remove precise amounts of patients fitted with contact lenses had diabetes [201]. Of the
corneal stromal tissue and the flap is then returned to its original practitioners surveyed, 55% felt that contact lens wear was safe for
position without sutures. In procedures such as epithelial laser in patients with diabetes, while 42% responded ‘maybe’ to the ques-
situ keratomileusis (Epi-LASIK), laser epithelial keratomileusis tion of safety and 3% felt that contact lens wear was not safe in
(LASEK), and photorefractive keratectomy (PRK), the laser is used to these patients [201]. The majority of practitioners felt that daily
refigure the curvature of the corneal surface of the cornea, hence wear was the appropriate modality with more regular aftercare
requiring removal of the epithelium. visits recommended for this patient group, compared to those
In the year 2000, the FDA declared diabetes to be a relative without diabetes [201].
contraindication to LASIK [191]. Patients with diabetes who un- The complications of contact lens use in patients with diabetes
dergo LASIK have been found to be at a significantly greater risk of have been reviewed by O'Donnell and Efron [26,202]. Studies on
post-operative corneal complications when compared to a group of diabetic contact lens complications tend to be in patients with
people without diabetes [192]. Fraunfelder and Rich first reported a advanced disease and when contact lenses are worn in the
complication rate of 47% in patients with diabetes compared to 6.9% extended wear modality [203,204]. To address the uncertainty
in those without diabetes, with the most common complications among practitioners, O'Donnell et al. conducted a prospective study
being erosions and persistent epithelial defects [192]. This study in which people with and without diabetes were fitted with contact
also reported poorer visual outcomes in the group with diabetes lenses for a 12-month period [205]. Over the observation period, no
[192]. The complications reported by Fraunfelder and Rich appear significant differences were found between the groups in terms of
to be related to the poor wound healing discussed above and may bulbar redness, corneal staining, corneal thickness, or corneal
be indicative of coexisting corneal neuropathy. Fraunfelder and sensitivity [205]. O'Donnell et al. reported no compromise to the
Rich, however, did not report on the severity of diabetes in their corneal integrity with contact lens wear by patients with diabetes
study. In a study by Halkiadakis et al. of 24 patients with well- [205], findings supported by March et al. [206] To establish the
controlled diabetes, 3 developed an epithelial defect post- effect of contact lens wear on corneal hydration in people with
operatively [193], indicating good short-term safety of LASIK in diabetes, O'Donnell and Efron fitted 23 people with and 23 people
diabetes. The age group in this study, however, was 10 years without diabetes with thick, low water content soft contact lenses
younger than that reported by Fraunfelder and Rich. In a study by and measured corneal thickness before and after contact lens wear
Cobo-Soriano and et al., a lower proportion of complications was [207]. They again found no difference in corneal thickness between
reported, with visual outcomes being no different than in the groups and no difference was found in the rate of recovery, leading
control group [194]. This study had a younger average age group these authors to conclude that the presence of diabetes should not
and a lower range of myopia than that reported by Fraunfelder and prevent clinicians from fitting their patients with diabetes with
Rich, which may explain the reported differences. A review by contact lenses [207]. Similarly, a 2011 paper found no difference in
Spadea and Paroli recommended that LASIK be considered in pa- the corneal thickness of people with diabetes, regardless of contact
tients with well-controlled diabetes and with no ocular manifes- lens wear status, although endothelial density was significantly less
tations of diabetes [195]. Patients with diabetes should be informed in the group wearing contact lenses [208].
of this risk and be monitored closely post-operatively. In the future, contact lenses may be increasingly used to detect
glucose levels in the tears, hence being useful as a means of diabetic
3.4.3. Other forms of ocular surgery metabolic control [150,209e212]. The available evidence indicates
Other forms of ocular surgery have also been associated with that with current materials, daily wear of contact lenses is safe in
complications when diabetes is a comorbidity. Trabeculectomy has people with diabetes. However, practitioners should be aware of
been associated with corneal epithelial defects, dellen, and fila- the impact that diabetes has on the ocular surface in general and
mentary keratitis [196]. The greatest risk factors for these compli- advise patients accordingly. Careful examination of the cornea and
cations post-trabeculectomy are pre-existing corneal staining and a tear film is needed during fitting and follow-up to identify prob-
history of diabetes [196]. These findings are in agreement with lems early.
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 53

4. The ocular surface microbiome in diabetes Acknowledgements

The combination of corneal neuropathy, poor wound healing, The authors wish to acknowledge Morag Stewart, Timothy Chan,
and dry eye places people with diabetes at increased risk of Jonathan Tolentino, Carmen Duong for their assistance with this
persistent epithelial defects [213], recurrent erosions [22,214,215], manuscript.
neurotrophic keratopathy [216], and possibly at higher risk of mi-
crobial keratitis [141,217e219].
Recent studies have suggested a role for the human gut micro- References
biome in the development of diabetes [220], with declines in Fir- [1] Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes
micutes and an increases in Bacteroidetes [221,222]. mellitus and its complications. Part 1: diagnosis and classification of diabetes
A study that investigated microbial diversity of the conjunctiva mellitus provisional report of a WHO consultation. Diabet Med 1998;15:
539e53.
in a rat model of streptozotocin-induced type 1 diabetes [223]
[2] Charnogursky G, Lee H, Lopez N. Diabetic neuropathy. Handb Clin Neurol
found that onset of diabetes was associated with reduced 2014;120:773e85.
complexity of the conjunctival microbiota, the disappearance of [3] International Diabetes Federation. IDF diabetes atlas. seventh ed. 2015.
species of staphylococci, Aerococcus viridans and Klebsiella pneu- http://www.diabetesatlas.org/.
[4] Seidell JC. Obesity, insulin resistance and diabetesea worldwide epidemic. Br
monia, but the appearance of enterococci, Kocuria kristinae, J Nutr 2000;83(Suppl 1):S5e8.
Enterobacter aerogenes, and Proteus vulgaris. [223] A study using a [5] WHO. Expert committee on diabetes mellitus. 1980.
streptozotocin mouse diabetes model showed that duration of [6] Tuomi T, Santoro N, Caprio S, Cai M, Weng J, Groop L. The many faces of
diabetes: a disease with increasing heterogeneity. Lancet 2014;383:
diabetes increased the risk of developing endogenous bacterial 1084e94.
endophthalmitis after injection of Klebsiella pneumoniae into the [7] Ginter E, Simko V. Type 2 diabetes mellitus, pandemic in 21st century. Adv
mouse tail vein [224]. The study also showed that longer duration Exp Med Biol 2012;771:42e50.
[8] World Health Organization, International Diabetes Federation. Definition and
of diabetes in these mice increased likelihood of developing diagnosis of diabetes mellitus and intermediate hyperglycemia: report of a
endophthalmitis [224]. WHO/IDF consultation. Geneva, Switzerland. 2006.
Most, but not all [225], studies have shown significant increases [9] Tuomi T, Groop LC, Zimmet PZ, Rowley MJ, Knowles W, Mackay IR. Anti-
bodies to glutamic acid decarboxylase reveal latent autoimmune diabetes
in microbes on the conjunctiva of diabetic patients. Habib et al. mellitus in adults with a non-insulin-dependent onset of disease. Diabetes
[226] found that there was an increase in bacterial diversity on the 1993;42:359e62.
conjunctiva with type 2, but not type 1, diabetes, and patients with [10] Kobayashi T, Tamemoto K, Nakanishi K, Kato N, Okubo M, Kajio H, et al.
Immunogenetic and clinical characterization of slowly progressive IDDM.
type 2 diabetes were significantly more commonly colonized by
Diabetes Care 1993;16:780e8.
Staphylococcus aureus [226]. Conjunctival swabs from people with [11] Colagiuri S. Epidemiology of prediabetes. Med Clin North Am 2011;95:
diabetes contain a greater frequency of coagulase negative staph- 299e307.
ylococci [227], Staphylococcus aureus, enterococci, certain strepto- [12] Atkinson MA. The pathogenesis and natural history of type 1 diabetes. Cold
Spring Harb Perspect Med 2012;2.
cocci, and Gram-negative bacteria including Klebsiella sp [228], [13] Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical
Enterobacter sp. and Escherichia coli [229,230]. Conjunctivas are interventions in type 1 diabetes. Nature 2010;464:1293e300.
more frequently colonized by microbes of patients with diabetic [14] Buysschaert M, Bergman M. Definition of prediabetes. Med Clin North Am
2011;95:289e97.
retinopathy [227,231]. The increased bacterial load on the con- [15] Cade WT. Diabetes-related microvascular and macrovascular diseases in the
junctiva may be related to severity of disease and perhaps to con- physical therapy setting. Phys Ther 2008;88:1322e35.
trol of glucose levels over time. [16] Alva ML, Gray A, Mihaylova B, Leal J, Holman RR. The impact of diabetes-
related complications on healthcare costs: new results from the UKPDS
(UKPDS 84). Diabet Med 2015;32:459e66.
5. Summary [17] Nowotny K, Jung T, Hohn A, Weber D, Grune T. Advanced glycation end
products and oxidative stress in type 2 diabetes mellitus. Biomolecules
2015;5:194e222.
Diabetic corneal neuropathy affects epithelial integrity, tear film
[18] Nunan D, Mahtani KR, Roberts N, Heneghan C. Physical activity for the
function, corneal sensitivity, and the healing response by reducing prevention and treatment of major chronic disease: an overview of sys-
the trophic support that is critical for ocular surface homeostasis. tematic reviews. Syst Rev 2013;2:56.
[19] Madden KM. Evidence for the benefit of exercise therapy in patients with
The severity of diabetic keratopathy can vary from dry eye symp-
type 2 diabetes. Diabetes Metab Syndr Obes 2013;6:233e9.
toms to visual impairment from corneal ulceration [26]. This review [20] Brownlee M. Biochemistry and molecular cell biology of diabetic complica-
highlights the need for clinical vigilance in detecting ocular surface tions. Nature 2001;414:813e20.
disease when examining patients with diabetes. [21] Sheetz MJ, King GL. Molecular understanding of hyperglycemia's adverse
effects for diabetic complications. JAMA 2002;288:2579e88.
Understanding the mechanisms resulting in the reduced corneal [22] Schultz RO, Van Horn DL, Peters MA, Klewin KM, Schutten WH. Diabetic
nerve density and the mechanisms contributing to the suscepti- keratopathy. Trans Am Ophthalmol Soc 1981;79:180e99.
bility to corneal erosion and infection in diabetes is critical, not only [23] Abdelkader H, Patel DV, McGhee C, Alany RG. New therapeutic approaches in
the treatment of diabetic keratopathy: a review. Clin Exp Ophthalmol
for prevention but also for insight into the systemic disease. The 2011;39:259e70.
cornea may be a simplistic model for understanding how diabetes [24] Didenko T, Smoliakova G, Sorokin E, Egorov V. Clinical and pathogenetic
affects other organs. There is a need to better characterize the features of neurotrophic corneal disorders in diabetes. Vestn Oftalmol
1999;115:7e11.
relationship between the neural, biochemical, and structural [25] Vieira-Potter VJ, Karamichos D, Lee DJ. Ocular complications of diabetes and
changes that occur in early diabetes. By understanding these re- therapeutic approaches. Biomed Res Int 2016;2016.
lationships, we can better prevent and predict the development of [26] O'Donnell C, Efron N. Diabetes and contact lens wear. Clin Exp Optom
2012;95:328e37.
diabetes-related complications. By modulating these changes, we [27] Sacchetti M, Lambiase A. Diagnosis and management of neurotrophic kera-
may be able, in the future, to slow down the progress of diabetic titis. Clin Ophthalmol 2014;8:571e9.
ocular surface disease and other diabetes-related complications. [28] Pritchard N, Edwards K, Russell AW, Perkins BA, Malik RA, Efron N. Corneal
confocal microscopy predicts 4-year incident peripheral neuropathy in type
1 diabetes. Diabetes Care 2015;38:671e5.
Funding [29] Cousen P, Cackett P, Bennett H, Swa K, Dhillon B. Tear production and corneal
sensitivity in diabetes. J Diabetes Complicat 2007;21:371e3.
No financial support was received for this work. [30] Dogru M, Katakami C, Inoue M. Tear function and ocular surface changes in
noninsulin-dependent diabetes mellitus. Ophthalmology 2001;108:586e92.
The authors have no proprietary or commercial interests in any [31] Friend J, Thoft RA. The diabetic cornea. Int Ophthalmol Clin 1984;24:111e23.
concept or product discussed in this article. [32] Yoon KC, Im SK, Seo MS. Changes of tear film and ocular surface in diabetes
54 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

mellitus. Korean J Ophthalmol 2004;18:168e74. Curr Drug Targets 2008;9:14e36.


[33] Pritchard N, Edwards K, Shahidi AM, Sampson GP, Russell AW, Malik RA, [63] Greene DA, Sima AA, Stevens MJ, Feldman EL, Lattimer SA. Complications:
et al. Corneal markers of diabetic neuropathy. Ocul Surf 2011;9:17e28. neuropathy, pathogenetic considerations. Diabetes Care 1992;15:1902e25.
[34] Schein OD, Glynn RJ, Poggio EC, Seddon JM, Kenyon KR. The relative risk of [64] Huang SP, Palla S, Ruzycki P, Varma RA, Harter T, Reddy GB, et al. Aldo-keto
ulcerative keratitis among users of daily-wear and extended-wear soft reductases in the eye. J Ophthalmol 2010;2010:521204.
contact lenses. A case-control study. Microbial Keratitis Study Group. N Engl J [65] Hempel A, Maasch C, Heintze U, Lindschau C, Dietz R, Luft FC, et al. High
Med 1989;321:773e8. glucose concentrations increase endothelial cell permeability via activation
[35] Patel DV, McGhee CN. Mapping of the normal human corneal sub-basal of protein kinase C alpha. Circ Res 1997;81:363e71.
nerve plexus by in vivo laser scanning confocal microscopy. Investig Oph- [66] Geraldes P, King GL. Activation of protein kinase C isoforms and its impact on
thalmol Vis Sci 2005;46:4485e8. diabetic complications. Circ Res 2010;106:1319e31.
[36] Muller LJ, Marfurt CF, Kruse F, Tervo TM. Corneal nerves: structure, contents [67] Obrosova IG. Diabetes and the peripheral nerve. Biochim Biophys Acta
and function. Exp Eye Res 2003;76:521e42. 2009;1792:931e40.
[37] Oliveira-Soto L, Efron N. Morphology of corneal nerves using confocal mi- [68] Farmer KL, Li C, Dobrowsky RT. Diabetic peripheral neuropathy: should a
croscopy. Cornea 2001;20:374e84. chaperone accompany our therapeutic approach? Pharmacol Rev 2012;64:
[38] Cai D, Zhu M, Petroll WM, Koppaka V, Robertson DM. The impact of type 1 880e900.
diabetes mellitus on corneal epithelial nerve morphology and the corneal [69] Lehning EJ, LoPachin RM, Mathew J, Eichberg J. Changes in Na-K ATPase and
epithelium. Am J Pathol 2014;184:2662e70. protein kinase C activities in peripheral nerve of acrylamide-treated rats.
[39] Misra SL, Craig JP, Patel DV, McGhee CN, Pradhan M, Ellyett K, et al. In vivo J Toxicol Environ Health 1994;42:331e42.
confocal microscopy of corneal nerves: an ocular biomarker for peripheral [70] Said G. Diabetic neuropathy-a review. Nat Clin Pract Neurol 2007;3:331e40.
and cardiac autonomic neuropathy in type 1 diabetes mellitus. Investig [71] Thomas PK. Classification, differential diagnosis, and staging of diabetic pe-
Ophthalmol Vis Sci 2015;56:5060e5. ripheral neuropathy. Diabetes 1997;46(Suppl 2):S54e7.
[40] Efron N. The Glenn A. Fry award lecture 2010: ophthalmic markers of dia- [72] Boulton AJ, Vileikyte L, Ragnarson-Tennvall G, Apelqvist J. The global burden
betic neuropathy. Optom Vis Sci 2011;88:661e83. of diabetic foot disease. Lancet 2005;366:1719e24.
[41] Cameron NE, Eaton SE, Cotter MA, Tesfaye S. Vascular factors and metabolic [73] Vinik AI, Mehrabyan A. Diabetic neuropathies. Med Clin North Am 2004;88:
interactions in the pathogenesis of diabetic neuropathy. Diabetologia 947e99.
2001;44:1973e88. [74] Vinik A, Ullal J, Parson HK, Casellini CM. Diabetic neuropathies: clinical
[42] Feldman EL, Stevens MJ, Greene DA. Pathogenesis of diabetic neuropathy. manifestations and current treatment options. Nat Clin Pract Endocrinol
Clin Neurosci 1997;4:365e70. Metab 2006;2:269e81.
[43] Ahsan H. Diabetic retinopathy-biomolecules and multiple pathophysiology. [75] Smith HS, Argoff CE. Pharmacological treatment of diabetic neuropathic pain.
Diabetes Metab Syndr 2015;9:51e4. Drugs 2011;71:557e89.
[44] Babizhayev MA, Strokov IA, Nosikov VV, Savel'yeva EL, Sitnikov VF, [76] Davies M, Brophy S, Williams R, Taylor A. The prevalence, severity, and
Yegorov YE, et al. The role of oxidative stress in diabetic neuropathy: gen- impact of painful diabetic peripheral neuropathy in type 2 diabetes. Diabetes
eration of free radical species in the glycation reaction and gene poly- Care 2006;29:1518e22.
morphisms encoding antioxidant enzymes to genetic susceptibility to [77] Pirart J. Diabetes mellitus and its degenerative complications: a prospective
diabetic neuropathy in population of type i diabetic patients. Cell Biochem study of 4,400 patients observed between 1947 and 1973. Diabete Metab
Biophys 2015;71:1425e43. 1977;3:97e107.
[45] Yagihashi S, Mizukami H, Sugimoto K. Mechanism of diabetic neuropathy: [78] Tavakoli M, Mitu-Pretorian M, Petropoulos IN, Fadavi H, Asghar O, Alam U,
where are we now and where to go? J Diabetes Investig 2011;2:18e32. et al. Corneal confocal microscopy detects early nerve regeneration in dia-
[46] Betteridge DJ. What is oxidative stress? Metabolism 2000;49:3e8. betic neuropathy after simultaneous pancreas and kidney transplantation.
[47] Duby JJ, Campbell RK, Setter SM, White JR, Rasmussen KA. Diabetic neu- Diabetes 2013;62:254e60.
ropathy: an intensive review. Am J Health Syst Pharm 2004;61:160e73. quiz [79] Albers JW, Brown MB, Sima AA, Greene DA. Nerve conduction measures in
75e76. mild diabetic neuropathy in the Early Diabetes Intervention Trial: the effects
[48] Vincent AM, Edwards JL, McLean LL, Hong Y, Cerri F, Lopez I, et al. Mito- of age, sex, type of diabetes, disease duration, and anthropometric factors.
chondrial biogenesis and fission in axons in cell culture and animal models of Tolrestat Study Group for the Early Diabetes Intervention Trial. Neurology
diabetic neuropathy. Acta Neuropathol 2010;120:477e89. 1996;46:85e91.
[49] Vincent AM, McLean LL, Backus C, Feldman EL. Short-term hyperglycemia [80] Sveen KA, Karime B, Jorum E, Mellgren SI, Fagerland MW, Monnier VM, et al.
produces oxidative damage and apoptosis in neurons. FASEB J 2005;19: Small- and large-fiber neuropathy after 40 years of type 1 diabetes: associ-
638e40. ations with glycemic control and advanced protein glycation: the Oslo Study.
[50] Hamid HS, Mervak CM, Munch AE, Robell NJ, Hayes JM, Porzio MT, et al. Diabetes Care 2013;36:3712e7.
Hyperglycemia- and neuropathy-induced changes in mitochondria within [81] Malik RA, Veves A, Tesfaye S, Smith G, Cameron N, Zochodne D, et al. Small
sensory nerves. Ann Clin Transl Neurol 2014;1:799e812. fibre neuropathy: role in the diagnosis of diabetic sensorimotor poly-
[51] Zherebitskaya E, Akude E, Smith DR, Fernyhough P. Development of selective neuropathy. Diabetes Metab Res Rev 2011;27:678e84.
axonopathy in adult sensory neurons isolated from diabetic rats: role of [82] Quattrini C, Tavakoli M, Jeziorska M, Kallinikos P, Tesfaye S, Finnigan J, et al.
glucose-induced oxidative stress. Diabetes 2009;58:1356e64. Surrogate markers of small fiber damage in human diabetic neuropathy.
[52] Tomlinson DR, Fernyhough P, Diemel LT. Role of neurotrophins in diabetic Diabetes 2007;56:2148e54.
neuropathy and treatment with nerve growth factors. Diabetes [83] Edwards K, Pritchard N, Vagenas D, Russell A, Malik RA, Efron N. Utility of
1997;46(Suppl 2):S43e9. corneal confocal microscopy for assessing mild diabetic neuropathy: baseline
[53] Kim J, Kim CS, Sohn E, Jeong IH, Kim H, Kim JS. Involvement of advanced findings of the LANDMark study. Clin Exp Optom 2012;95:348e54.
glycation end products, oxidative stress and nuclear factor-kappaB in the [84] Perry JR, Bril V. Complications of sural nerve biopsy in diabetic versus non-
development of diabetic keratopathy. Graefes Arch Clin Exp Ophthalmol diabetic patients. Can J Neurol Sci 1994;21:34e7.
2011;249:529e36. [85] Tavakoli M, Quattrini C, Abbott C, Kallinikos P, Marshall A, Finnigan J, et al.
[54] Aubert CE, Michel PL, Gillery P, Jaisson S, Fonfrede M, Morel F, et al. Asso- Corneal confocal microscopy: a novel noninvasive test to diagnose and
ciation of peripheral neuropathy with circulating advanced glycation end stratify the severity of human diabetic neuropathy. Diabetes Care 2010;33:
products, soluble receptor for advanced glycation end products and other 1792e7.
risk factors in patients with type 2 diabetes. Diabetes Metab Res Rev [86] Dyck PJ, Norell JE, Tritschler H, Schuette K, Samigullin R, Ziegler D, et al.
2014;30:679e85. Challenges in design of multicenter trials: end points assessed longitudinally
[55] Ryle C, Donaghy M. Non-enzymatic glycation of peripheral nerve proteins in for change and monotonicity. Diabetes Care 2007;30:2619e25.
human diabetics. J Neurol Sci 1995;129:62e8. [87] Dehghani C, Pritchard N, Edwards K, Vagenas D, Russell AW, Malik RA, et al.
[56] Brownlee M, Vlassara H, Cerami A. Nonenzymatic glycosylation and the Natural history of corneal nerve morphology in mild neuropathy associated
pathogenesis of diabetic complications. Ann Intern Med 1984;101:527e37. with type 1 diabetes: development of a potential measure of diabetic pe-
[57] Sugimoto K, Nishizawa Y, Horiuchi S, Yagihashi S. Localization in human ripheral neuropathy. Investig Ophthalmol Vis Sci 2014;55:7982e90.
diabetic peripheral nerve of N ε -carboxymethyllysine-protein adducts, an [88] Mehra S, Tavakoli M, Kallinikos PA, Efron N, Boulton AJ, Augustine T, et al.
advanced glycation endproduct. Diabetologia 1997;40:1380e7. Corneal confocal microscopy detects early nerve regeneration after pancreas
[58] Sugimoto K, Yasujima M, Yagihashi S. Role of advanced glycation end transplantation in patients with type 1 diabetes. Diabetes Care 2007;30:
products in diabetic neuropathy. Curr Pharm Des 2008;14:953e61. 2608e12.
[59] McDermott AM, Xiao TL, Kern TS, Murphy CJ. Non-enzymatic glycation in [89] Ziegler D, Papanas N, Zhivov A, Allgeier S, Winter K, Ziegler I, et al. Early
corneas from normal and diabetic donors and its effects on epithelial cell detection of nerve fiber loss by corneal confocal microscopy and skin biopsy
attachment in vitro. Optom St Louis Mo 2003;74:443e52. in recently diagnosed type 2 diabetes. Diabetes 2014;63:2454e63.
[60] Stavniichuk R, Shevalye H, Hirooka H, Nadler JL, Obrosova IG. Interplay of [90] Dehghani C, Pritchard N, Edwards K, Vagenas D, Russell AW, Malik RA, et al.
sorbitol pathway of glucose metabolism, 12/15-lipoxygenase, and mitogen- Morphometric stability of the corneal subbasal nerve plexus in healthy in-
activated protein kinases in the pathogenesis of diabetic peripheral neu- dividuals: a 3-year longitudinal study using corneal confocal microscopy.
ropathy. Biochem Pharmacol 2012;83:932e40. Investig Ophthalmol Vis Sci 2014;55:3195e9.
[61] Chung SS, Ho EC, Lam KS, Chung SK. Contribution of polyol pathway to [91] Hu L, Chen J, Zhang L, Sun X, Huang J, Xie W, et al. Effects of long-term soft
diabetes-induced oxidative stress. J Am Soc Nephrol 2003;14:S233e6. contact lenses on tear menisci and corneal nerve density. Eye Contact Lens
[62] Oates PJ. Aldose reductase, still a compelling target for diabetic neuropathy. 2016;42:196e201.
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 55

[92] Niederer RL, Perumal D, Sherwin T, McGhee CN. Age-related differences in [122] Nishida T, Nakamura M, Ofuji K, Reid TW, Mannis MJ, Murphy CJ. Synergistic
the normal human cornea: a laser scanning in vivo confocal microscopy effects of substance P with insulin-like growth factor-1 on epithelial
study. Br J Ophthalmol 2007;91:1165e9. migration of the cornea. J Cell Physiol 1996;169:159e66.
[93] Parissi M, Karanis G, Randjelovic S, Germundsson J, Poletti E, Ruggeri A, et al. [123] Nakamura M, Ofuji K, Chikama T, Nishida T. Combined effects of substance P
Standardized baseline human corneal subbasal nerve density for clinical and insulin-like growth factor-1 on corneal epithelial wound closure of
investigations with laser-scanning in vivo confocal microscopy. Investig rabbit in vivo. Curr Eye Res 1997;16:275e8.
Ophthalmol Vis Sci 2013;54:7091e102. [124] Nakamura M, Chikama T, Nishida T. Up-regulation of integrin alpha 5
[94] Cruzat A, Qazi Y, Hamrah P. In vivo confocal microscopy of corneal nerves in expression by combination of substance P and insulin-like growth factor-1 in
health and disease. Ocul Surf 2017;15:15e47. rabbit corneal epithelial cells. Biochem Biophys Res Commun 1998;246:
[95] Petropoulos IN, Alam U, Fadavi H, Asghar O, Green P, Ponirakis G, et al. 777e82.
Corneal nerve loss detected with corneal confocal microscopy is symmetrical [125] Reid TW, Murphy CJ, Iwahashi CK, Foster BA, Mannis MJ. Stimulation of
and related to the severity of diabetic polyneuropathy. Diabetes Care epithelial cell growth by the neuropeptide substance P. J Cell Biochem
2013;36:3646e51. 1993;52:476e85.
[96] Pritchard N, Edwards K, Dehghani C, Fadavi H, Jeziorska M, Marshall A, et al. [126] Garcia-Hirschfeld J, Lopez-Briones LG, Belmonte C. Neurotrophic influences
Longitudinal assessment of neuropathy in type 1 diabetes using novel on corneal epithelial cells. Exp Eye Res 1994;59:597e605.
ophthalmic markers (LANDMark): study design and baseline characteristics. [127] Kunt T, Forst T, Schmidt S, Pfutzner A, Schneider S, Harzer O, et al. Serum
Diabetes Res Clin Pract 2014;104:248e56. levels of substance P are decreased in patients with type 1 diabetes. Exp Clin
[97] Jiang MS, Yuan Y, Gu ZX, Zhuang SL. Corneal confocal microscopy for Endocrinol Diabetes 2000;108:164e7.
assessment of diabetic peripheral neuropathy: a meta-analysis. Br J Oph- [128] Stapleton F, Chao C, Jalbert I, Mandathara PS, Kolanu S, Willcox MDP, et al.
thalmol 2016;100:9e14. Relationships between central corneal nerve parameters and tear neuro-
[98] Leppin K, Behrendt AK, Reichard M, Stachs O, Guthoff RF, Baltrusch S, et al. peptides. Investig Ophthalmol Vis Sci 2016;57.
Diabetes mellitus leads to accumulation of dendritic cells and nerve fiber [129] Markoulli M, Lum E, You JJ, Duong C, Tolentino J, Kim J. Corneal nerve
damage of the subbasal nerve plexus in the cornea. Investig Ophthalmol Vis morphology and tear film substance p in diabetes. Investig Ophthalmol Vis
Sci 2014;55:3603e15. Sci 2016;57:6153.
[99] Edwards K, Pritchard N, Poole C, Dehghani C, Al Rashah K, Russell A, et al. [130] Yamada M, Ogata M, Kawai M, Mashima Y. Decreased substance P concen-
Development of a novel technique to measure corneal nerve migration rate. trations in tears from patients with corneal hypesthesia. Am J Ophthalmol
Cornea 2016;35:700e5. 2000;129:671e2.
[100] Tavakoli M, Kallinikos P, Iqbal A, Herbert A, Fadavi H, Efron N, et al. Corneal [131] Yamada M, Ogata M, Kawai M, Mochizuki H, Mashima Y. Topical diclofenac
confocal microscopy detects improvement in corneal nerve morphology sodium decreases the substance P content of tears. Arch Ophthalmol
with an improvement in risk factors for diabetic neuropathy. Diabet Med 2002;120:51e4.
2011;28:1261e7. [132] Cashman JN. The mechanisms of action of NSAIDs in analgesia. Drugs
[101] Smith AG, Russell J, Feldman EL, Goldstein J, Peltier A, Smith S, et al. Lifestyle 1996;52(Suppl 5):13e23.
intervention for pre-diabetic neuropathy. Diabetes Care 2006;29:1294e9. [133] Mohamed-Noriega K, Butron-Valdez K, Vazquez-Galvan J, Mohamed-
[102] Shaheen BS, Bakir M, Jain S. Corneal nerves in health and disease. Surv Noriega J, Cavazos-Adame H, Mohamed-Hamsho J. Corneal melting after
Ophthalmol 2014;59:263e85. collagen cross-linking for keratoconus in a thin cornea of a diabetic patient
[103] Hsu HY, Modi D. Etiologies, quantitative hypoesthesia, and clinical outcomes treated with topical nepafenac: a case report with a literature review. Case
of neurotrophic keratopathy. Eye Contact Lens 2015;41:314e7. Rep Ophthalmol 2016;7:119e24.
[104] Shih KC, Lam KSL, Tong L. A systematic review on the impact of diabetes [134] Marfurt CF, Echtenkamp SF. The effect of diabetes on neuropeptide content
mellitus on the ocular surface. Nutr Diabetes 2017;7. e251. in the rat cornea and iris. Investig Ophthalmol Vis Sci 1995;36:1100e6.
[105] Yeh S, Song XJ, Farley W, Li DQ, Stern ME, Pflugfelder SC. Apoptosis of ocular [135] Bonini S, Lambiase A, Rama P, Caprioglio G, Aloe L. Topical treatment with
surface cells in experimentally induced dry eye. Investig Ophthalmol Vis Sci nerve growth factor for neurotrophic keratitis. Ophthalmology 2000;107:
2003;44:124e9. 1347e51. discussion 51e52.
[106] Gekka M, Miyata K, Nagai Y, Nemoto S, Sameshima T, Tanabe T, et al. Corneal [136] Lambiase A, Bonini S, Micera A, Rama P, Bonini S, Aloe L. Expression of nerve
epithelial barrier function in diabetic patients. Cornea 2004;23:35e7. growth factor receptors on the ocular surface in healthy subjects and during
[107] Chang SW, Hsu HC, Hu FR, Chen MS. Corneal autofluorescence and epithelial manifestation of inflammatory diseases. Investig Ophthalmol Vis Sci
barrier function in diabetic patients. Ophthalmic Res 1995;27:74e9. 1998;39:1272e5.
[108] Alper M. The anesthetic eye: an investigation of changes in the anterior [137] Lambiase A, Micera A, Pellegrini G, Merlo D, Rama P, De Luca M, et al. In vitro
ocular segment of the monkey caused by interrupting the trigeminal nerve evidence of nerve growth factor effects on human conjunctival epithelial cell
at various levels along its course. Trans Am Ophthalmol Soc 1975;73:313. differentiation and mucin gene expression. Investig Ophthalmol Vis Sci
[109] Mackie I. Role of the corneal nerves in destructive disease of the cornea. 2009;50:4622e30.
Trans Ophthalmol Soc U. K 1978;98:343. [138] Rocha EM, Cunha DA, Carneiro EM, Boschero AC, Saad MJ, Velloso LA.
[110] Beuerman RW, Schimmelpfennig B. Sensory denervation of the rabbit cornea Identification of insulin in the tear film and insulin receptor and IGF-1 re-
affects epithelial properties. Exp Neurol 1980;69:196e201. ceptor on the human ocular surface. Investig Ophthalmol Vis Sci 2002;43:
[111] Yin J, Huang J, Chen C, Gao N, Wang F, Yu FS. Corneal complications in 963e7.
streptozocin-induced type I diabetic rats. Investig Ophthalmol Vis Sci [139] Cunha DA, de Alves MC, Stoppiglia LF, Jorge AG, Modulo CM, Carneiro EM,
2011;52:6589e96. et al. Extra-pancreatic insulin production in RAt lachrymal gland after
[112] Kaji Y, Usui T, Oshika T, Matsubara M, Yamashita H, Araie M, et al. Advanced streptozotocin-induced islet beta-cells destruction. Biochim Biophys Acta
glycation end products in diabetic corneas. Investig Ophthalmol Vis Sci 2007;1770:1128e35.
2000;41:362e8. [140] Ding J, Liu Y, Sullivan DA. Effects of insulin and high glucose on human
[113] Xu KP, Li Y, Ljubimov AV, Yu FS. High glucose suppresses epidermal growth meibomian gland epithelial cells. Investig Ophthalmol Vis Sci 2015;56:
factor receptor/phosphatidylinositol 3-kinase/Akt signaling pathway and 7814e20.
attenuates corneal epithelial wound healing. Diabetes 2009;58:1077e85. [141] Alves Mde C, Carvalheira JB, Modulo CM, Rocha EM. Tear film and ocular
[114] Kim SJ, Toma HS. Ophthalmic antibiotics and antimicrobial resistance a surface changes in diabetes mellitus. Arq Bras Oftalmol 2008;71:96e103.
randomized, controlled study of patients undergoing intravitreal injections. [142] Cruzat A, Pavan-Langston D, Hamrah P. In vivo confocal microscopy of
Ophthalmology 2011;118:1358e63. corneal nerves: analysis and clinical correlation. Semin Ophthalmol 2010;25:
[115] Nakamura M, Kawahara M, Nakata K, Nishida T. Restoration of corneal 171e7.
epithelial barrier function and wound healing by substance P and IGF-1 in [143] Chen DK, Frizzi KE, Guernsey LS, Ladt K, Mizisin AP, Calcutt NA. Repeated
rats with capsaicin-induced neurotrophic keratopathy. Investig Ophthalmol monitoring of corneal nerves by confocal microscopy as an index of pe-
Vis Sci 2003;44:2937e40. ripheral neuropathy in type-1 diabetic rodents and the effects of topical
[116] Ljubimov AV. Diabetic complications in the cornea. Vis Res 2017 Apr 28. insulin. J Peripher Nerv Syst 2013;18:306e15.
https://doi.org/10.1016/j.visres.2017.03.002. pii: S0042e6989(17)30047- [144] Zagon IS, Klocek MS, Sassani JW, McLaughlin PJ. Use of topical insulin to
0 [Epub ahead of print]. normalize corneal epithelial healing in diabetes mellitus. Arch Ophthalmol
[117] Lambiase A, Micera A, Sacchetti M, Cortes M, Mantelli F, Bonini S. Alterations 2007;125:1082e8.
of tear neuromediators in dry eye disease. Arch Ophthalmol 2011;129: [145] Zagon IS, Sassani JW, McLaughlin PJ. Insulin treatment ameliorates impaired
981e6. corneal reepithelialization in diabetic rats. Diabetes 2006;55:1141e7.
[118] Yamada M, Ogata M, Kawai M, Mashima Y, Nishida T. Substance P in human [146] Klocek MS, Sassani JW, McLaughlin PJ, Zagon IS. Naltrexone and insulin are
tears. Cornea 2003;22:S48e54. independently effective but not additive in accelerating corneal epithelial
[119] Felipe CD, Gonzalez GG, Gallar J, Belmonte C. Quantification and immuno- healing in type I diabetic rats. Exp Eye Res 2009;89:686e92.
cytochemical characteristics of trigeminal ganglion neurons projecting to the [147] Wu YC, Buckner BR, Zhu M, Cavanagh HD, Robertson DM. Elevated IGFBP3
cornea: effect of corneal wounding. Eur J Pain 1999;3:31e9. levels in diabetic tears: a negative regulator of IGF-1 signaling in the corneal
[120] Stern ME, Pflugfelder SC. Inflammation in dry eye. Ocul Surf 2004;2:124e30. epithelium. Ocul Surf 2012;10:100e7.
[121] Nakamura M, Chikama T, Nishida T. Synergistic effect with Phe-Gly-Leu- [148] Gasset AR, Braverman LE, Fleming MC, Arky RA, Alter BR. Tear glucose
Met-NH2 of the C-terminal of substance P and insulin-like growth factor-1 detection of hyperglycemia. Am J Ophthalmol 1968;65:414e20.
on epithelial wound healing of rabbit cornea. Br J Pharmacol 1999;127: [149] Sen DK, Sarin GS. Tear glucose levels in normal people and in diabetic pa-
489e97. tients. Br J Ophthalmol 1980;64:693e5.
56 M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57

[150] Peng B, Lu J, Balijepalli AS, Major TC, Cohan BE, Meyerhoff ME. Evaluation of [182] Javitt JC, Street DA, Tielsch JM, Wang Q, Kolb MM, Schien O, et al. National
enzyme-based tear glucose electrochemical sensors over a wide range of outcomes of cataract extraction. Retinal detachment and endophthalmitis
blood glucose concentrations. Biosens Bioelectron 2013;49:204e9. after outpatient cataract surgery. Cataract Patient Outcomes Research Team.
[151] Zhao Z, Liu J, Shi B, He S, Yao X, Willcox MD. Advanced glycation end product Ophthalmology 1994;101:100e5. discussion 6.
modified proteins in tears of diabetic patients. Mol Vis 2010;16:1576e84. [183] Menikoff JA, Speaker MG, Marmor M, Raskin EM. A case-control study of risk
[152] Grus FH, Sabuncuo P, Dick HB, Augustin AJ, Pfeiffer N. Changes in the tear factors for postoperative endophthalmitis. Ophthalmology 1991;98:1761e8.
proteins of diabetic patients. BMC Ophthalmol 2002;2:4. [184] Misra SL, Goh YW, Patel DV, Riley AF, McGhee CN. Corneal microstructural
[153] Stolwijk TR, Kuizenga A, van Haeringen NJ, Kijlstra A, Oosterhuis JA, van changes in nerve fiber, endothelial and epithelial density after cataract
Best JA. Analysis of tear fluid proteins in insulin-dependent diabetes melli- surgery in patients with diabetes mellitus. Cornea 2015;34:177e81.
tus. Acta Ophthalmol Copenh 1994;72:357e62. [185] Jiang D, Xiao X, Fu T, Mashaghi A, Liu Q, Hong J. Transient tear film
[154] de Luca C, Olefsky JM. Inflammation and insulin resistance. FEBS Lett dysfunction after cataract surgery in diabetic patients. PloS One 2016;11,
2008;582:97e105. e0146752.
[155] Costagliola C, Romano V, De Tollis M, Aceto F, dell'Omo R, Romano MR, et al. [186] Gemensky-Metzler AJ, Sheahan JE, Rajala-Schultz PJ, Wilkie DA, Harrington J.
TNF-alpha levels in tears: a novel biomarker to assess the degree of diabetic Retrospective study of the prevalence of keratoconjunctivitis sicca in dia-
retinopathy. Mediat Inflamm 2013;2013:629529. betic and nondiabetic dogs after phacoemulsification. Vet Ophthalmol
[156] Gill SE, Parks WC. Metalloproteinases and their inhibitors: regulators of 2015;18:472e80.
wound healing. Int J Biochem Cell Biol 2008;40:1334e47. [187] Chikamoto N, Chikama T, Yamada N, Nishida T, Ishimitsu T, Kamiya A. Effi-
[157] Nagase H, Woessner Jr JF. Matrix metalloproteinases. J Biol Chem 1999;274: cacy of substance P and insulin-like growth factor-1 peptides for preventing
21491e4. postsurgical superficial punctate keratopathy in diabetic patients. Jpn J
[158] Berman M, Dohlman CH, Gnadinger M, Davison P. Characterization of col- Ophthalmol 2009;53:464e9.
lagenolytic activity in the ulcerating cornea. Exp Eye Res 1971;11:255e7. [188] Schulze SD, Sekundo W, Kroll P. Autologous serum for the treatment of
[159] Garrana RM, Zieske JD, Assouline M, Gipson IK. Matrix metalloproteinases in corneal epithelial abrasions in diabetic patients undergoing vitrectomy. Am J
epithelia from human recurrent corneal erosion. Investig Ophthalmol Vis Sci Ophthalmol 2006;142:207e11.
1999;40:1266e70. [189] Fujishima H, Tsubota K. Improvement of corneal fluorescein staining in post
[160] Schultz GS, Wysocki A. Interactions between extracellular matrix and cataract surgery of diabetic patients by an oral aldose reductase inhibitor,
growth factors in wound healing. Wound Repair Regen 2009;17:153e62. ONO-2235. Br J Ophthalmol 2002;86:860e3.
[161] Kaiserman I, Kaiserman N, Nakar S, Vinker S. Dry eye in diabetic patients. Am [190] Nakahara M, Miyata K, Otani S, Miyai T, Nejima R, Yamagami S, et al.
J Ophthalmol 2005;139:498e503. A randomised, placebo controlled clinical trial of the aldose reductase in-
[162] Lv H, Li A, Zhang X, Xu M, Qiao Y, Zhang J, et al. Meta-analysis and review on hibitor CT-112 as management of corneal epithelial disorders in diabetic
the changes of tear function and corneal sensitivity in diabetic patients. Acta patients. Br J Ophthalmol 2005;89:266e8.
Ophthalmol 2014;92:e96e104. [191] United States Food and Drug Administration. When is LASIK not for me?
[163] Seifart U, Strempel I. The dry eye and diabetes mellitus. Ophthalmologe http://www.fda.gov/medicaldevices/productsandmedicalprocedures/
1994;91:235e9. surgeryandlifesupport/lasik/ucm061366.htm2014.
[164] Creuzot-Garcher C, Lafontaine PO, Gualino O, D'Athis P, Petit JM, Bron A. [192] Fraunfelder FW, Rich LF. Laser-assisted in situ keratomileusis complications
Study of ocular surface involvement in diabetic patients. J Fr Ophtalmol in diabetes mellitus. Cornea 2002;21:246e8.
2005;28:583e8. [193] Halkiadakis I, Belfair N, Gimbel HV. Laser in situ keratomileusis in patients
[165] Goebbels M. Tear secretion and tear film function in insulin dependent di- with diabetes. J Cataract Refract Surg 2005;31:1895e8.
abetics. Br J Ophthalmol 2000;84:19e21. [194] Cobo-Soriano R, Beltran J, Baviera J. LASIK outcomes in patients with un-
[166] Sullivan DA, Edwards JA, Wickham LA, Pena JD, Gao J, Ono M, et al. Identi- derlying systemic contraindications: a preliminary study. Ophthalmology
fication and endocrine control of sex steroid binding sites in the lacrimal 2006;113(1118):e1e8.
gland. Curr Eye Res 1996;15:279e91. [195] Spadea L, Paroli MP. Laser refractive surgery in diabetic patients: a review of
[167] Shetty R, Saeed T, Rashed H, Adeghate E, Singh J. Effect of diabetes mellitus the literature. Clin Ophthalmol 2012;6:1775e83.
on acinar morphology, peroxidase concentration, and release in isolated rat [196] Ono T, Yuki K, Ozeki N, Shiba D, Tsubota K. Ocular surface complications after
lacrimal glands. Curr Eye Res 2009;34:905e11. trabeculectomy: incidence, risk factors, time course and prognosis. Oph-
[168] Inoue K, Kato S, Ohara C, Numaga J, Amano S, Oshika T. Ocular and systemic thalmologica 2013;230:93e9.
factors relevant to diabetic keratoepitheliopathy. Cornea 2001;20:798e801. [197] Friberg TR, Ohji M, Scherer JJ, Tano Y. Frequency of epithelial debridement
[169] Sagdik HM, Ugurbas SH, Can M, Tetikoglu M, Ugurbas E, Ugurbas SC, et al. during diabetic vitrectomy. Am J Ophthalmol 2003;135:553e4.
Tear film osmolarity in patients with diabetes mellitus. Ophthalmic Res [198] Shimada H, Nakashizuka H, Hattori T, Mori R, Mizutani Y, Yuzawa M. Inci-
2013;50:1e5. dence of endophthalmitis after 20- and 25-gauge vitrectomy causes and
[170] DeMill DL, Hussain M, Pop-Busui R, Shtein RM. Ocular surface disease in prevention. Ophthalmology 2008;115:2215e20.
patients with diabetic peripheral neuropathy. Br J Ophthalmol 2016;100: [199] Dogru M, Kaderli B, Gelisken O, Yucel A, Avci R, Goto E, et al. Ocular surface
924e8. changes with applanation contact lens and coupling fluid use after argon
[171] Beckman KA. Characterization of dry eye disease in diabetic patients versus laser photocoagulation in noninsulin-dependent diabetes mellitus. Am J
nondiabetic patients. Cornea 2014;33:851e4. Ophthalmol 2004;138:381e8.
[172] Leibowitz HM, Krueger DE, Maunder LR, Milton RC, Kini MM, Kahn HA, et al. [200] Lu L, Reinach PS, Kao WW. Corneal epithelial wound healing. Exp Biol Med
The Framingham Eye Study monograph: an ophthalmological and epide- Maywood 2001;226:653e64.
miological study of cataract, glaucoma, diabetic retinopathy, macular [201] Veys J, Efron N, Boulton A. A survey of contact lens wear among diabetic
degeneration, and visual acuity in a general population of 2631 adults, 1973- patients in the United Kingdom. Cont Lens Anterior Eye 1997;20:S27e33.
1975. Surv Ophthalmol 1980;24:335e610. [202] O'Donnell C, Efron N. Contact lens wear and diabetes mellitus. Cont Lens
[173] Ederer F, Hiller R, Taylor HR. Senile lens changes and diabetes in two pop- Anterior Eye 1998;21:19e26.
ulation studies. Am J Ophthalmol 1981;91:381e95. [203] Eichenbaum JW, Feldstein M, Podos SM. Extended-wear aphakic soft contact
[174] Haddad NM, Sun JK, Abujaber S, Schlossman DK, Silva PS. Cataract surgery lenses and corneal ulcers. Br J Ophthalmol 1982;66:663e6.
and its complications in diabetic patients. Semin Ophthalmol 2014;29: [204] Spoor TC, Hartel WC, Wynn P, Spoor DK. Complications of continuous-wear
329e37. soft contact lenses in a nonreferral population. Arch Ophthalmol 1984;102:
[175] Pershing S, Morrison DE, Hernandez-Boussard T. Cataract surgery compli- 1312e3.
cations and revisit rates among three states. Am J Ophthalmol 2016;171: [205] O'Donnell C, Efron N, Boulton AJ. A prospective study of contact lens wear in
130e8. diabetes mellitus. Ophthalmic Physiol Opt 2001;21:127e38.
[176] Ozdemir M, Buyukbese MA, Cetinkaya A, Ozdemir G. Risk factors for ocular [206] March W, Long B, Hofmann W, Keys D, McKenney C. Safety of contact lenses
surface disorders in patients with diabetes mellitus. Diabetes Res Clin Pract in patients with diabetes. Diabetes Technol Ther 2004;6:49e52.
2003;59:195e9. [207] O'Donnell C, Efron N. Corneal hydration control in contact lens wearers with
[177] Tsaousis KT, Panagiotou DZ, Kostopoulou E, Vlatsios V, Stampouli D. Corneal diabetes mellitus. Optom Vis Sci 2006;83:22e6.
oedema after phacoemulsification in the early postoperative period: a [208] Leem HS, Lee KJ, Shin KC. Central corneal thickness and corneal endothelial
qualitative comparative case-control study between diabetics and non-di- cell changes caused by contact lens use in diabetic patients. Yonsei Med J
abetics. Ann Med Surg Lond 2016;5:67e71. 2011;52:322e5.
[178] Hugod M, Storr-Paulsen A, Norregaard JC, Nicolini J, Larsen AB, Thulesen J. [209] March WF, Mueller A, Herbrechtsmeier P. Clinical trial of a noninvasive
Corneal endothelial cell changes associated with cataract surgery in patients contact lens glucose sensor. Diabetes Technol Ther 2004;6:782e9.
with type 2 diabetes mellitus. Cornea 2011;30:749e53. [210] Phan CM, Subbaraman L, Jones LW. The use of contact lenses as biosensors.
[179] Mathew PT, David S, Thomas N. Endothelial cell loss and central corneal Optom Vis Sci 2016;93:419e25.
thickness in patients with and without diabetes after manual small incision [211] Ascaso FJ, Huerva V. Noninvasive continuous monitoring of tear glucose
cataract surgery. Cornea 2011;30:424e8. using glucose-sensing contact lenses. Optom Vis Sci 2016;93:426e34.
[180] Kattan HM, Flynn Jr HW, Pflugfelder SC, Robertson C, Forster RK. Nosocomial [212] Farandos NM, Yetisen AK, Monteiro MJ, Lowe CR, Yun SH. Contact lens
endophthalmitis survey. Current incidence of infection after intraocular sensors in ocular diagnostics. Adv Healthc Mater 2015;4:792e810.
surgery. Ophthalmology 1991;98:227e38. [213] Azar DT, Spurrmichaud SJ, Tisdale AS, Gipson IK. Decreased penetration of
[181] Phillips 2nd WB, Tasman WS. Postoperative endophthalmitis in association anchoring fibrils into the diabetic stroma - a morphometric analysis. Arch
with diabetes mellitus. Ophthalmology 1994;101:508e18. Ophthalmol 1989;107:1520e3.
M. Markoulli et al. / The Ocular Surface 16 (2018) 45e57 57

[214] Dursun D, Kim MC, Solomon A, Pflugfelder SC. Treatment of recalcitrant differs from non-diabetic adults. PloS One 2010;5, e9085.
recurrent corneal erosions with inhibitors of matrix metalloproteinase-9, [223] Yang C, Fei Y, Qin Y, Luo D, Yang S, Kou X, et al. Bacterial flora changes in
doxycycline and corticosteroids. Am J Ophthalmol 2001;132:8e13. conjunctiva of rats with streptozotocin-induced type i diabetes. PloS One
[215] Akpek EK, Merchant A, Pinar V, Foster CS. Ocular rosacea: patient charac- 2015;10, e0133021.
teristics and follow-up. Ophthalmology 1997;104:1863e7. [224] Coburn PS, Wiskur BJ, Christy E, Callegan MC. The diabetic ocular environ-
[216] Bonini S, Rama P, Olzi D, Lambiase A. Neurotrophic keratitis. Eye Lond ment facilitates the development of endogenous bacterial endophthalmitis.
2003;17:989e95. Investig Ophthalmol Vis Sci 2012;53:7426e31.
[217] Cho BJ, Lee GJ, Ha SY, Seo YH, Tchah H. Co-infection of the human cornea [225] Moreno NP, Moreno RD, Sousa LB. Aerobic bacterial microbiota of the con-
with Stenotrophomonas maltophilia and Aspergillus fumigatus. Cornea junctiva in diabetic patients with normal and altered glycated hemoglobin
2002;21:628e31. levels in two regions in Brazil. Arq Bras Oftalmol 2014;77:351e4.
[218] Holifield K, Lazzaro DR. Case report: spontaneous Stenotrophomonas mal- [226] Bilen H, Ates O, Astam N, Uslu H, Akcay G, Baykal O. Conjunctival flora in
tophilia keratitis in a diabetic patient. Eye Contact Lens 2011;37:326e7. patients with type 1 or type 2 diabetes mellitus. Adv Ther 2007;24:1028e35.
[219] Jin H, Parker WT, Law NW, Clarke CL, Gisseman JD, Pflugfelder SC, et al. [227] Karimsab D, Razak SK. Study of aerobic bacterial conjunctival flora in pa-
Evolving risk factors and antibiotic sensitivity patterns for microbial keratitis tients with diabetes mellitus. Nepal J Ophthalmol 2013;5:28e32.
at a large county hospital. Br J Ophthalmol 2017;101(11):1483e7. [228] Fernandez-Rubio ME, Rebolledo-Lara L, Martinez-Garcia M, Alarcon-
[220] Brown CT, Davis-Richardson AG, Giongo A, Gano KA, Crabb DB, Mukherjee N, Tomas M, Cortes-Valdes C. The conjunctival bacterial pattern of diabetics
et al. Gut microbiome metagenomics analysis suggests a functional model undergoing cataract surgery. Eye Lond 2010;24:825e34.
for the development of autoimmunity for type 1 diabetes. PloS One 2011;6, [229] Nahar N, Anwar S, Miah MRA. Conjunctival bacterial flora in diabetic pa-
e25792. tients. Ibrahim Med Coll J 2013;7:4.
[221] Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, et al. To- [230] Adam M, Balci M, Bayhan HA, Inkaya AC, Uyar M, Gurdal C. Conjunctival flora
ward defining the autoimmune microbiome for type 1 diabetes. ISME J in diabetic and nondiabetic individuals. Turk J Ophthalmol 2015;45:193e6.
2011;5:82e91. [231] Martins EN, Alvarenga LS, Hofling-Lima AL, Freitas D, Zorat-Yu MC, Farah ME,
[222] Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, et al. Aerobic bacterial conjunctival flora in diabetic patients. Cornea
Pedersen BK, et al. Gut microbiota in human adults with type 2 diabetes 2004;23:136e42.

Você também pode gostar