ArticlePDF AvailableLiterature Review
Progress in Retinal and Eye Research 97 (2023) 101217
Available online 30 September 2023
1350-9462/© 2023 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
Neurovascular dysfunction in glaucoma
Luis Alarcon-Martinez
a
,
b
,
c
, Yukihiro Shiga
a
,
b
, Deborah Villafranca-Baughman
a
,
b
,
Jorge L. Cueva Vargas
a
,
b
, Isaac A. Vidal Paredes
a
,
b
, Heberto Quintero
a
,
b
, Brad Fortune
d
,
Helen Danesh-Meyer
e
, Adriana Di Polo
a
,
b
,
*
a
Department of Neuroscience, Universit´
e de Montr´
eal, PO Box 6128, Station centre-ville, Montreal, QC, Canada
b
Neuroscience Division, Centre de recherche du Centre Hospitalier de lUniversit´
e de Montr´
eal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
c
Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
d
Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
e
Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
ARTICLE INFO
Keywords:
Retinal ganglion cells
Glaucoma
Neurovascular unit
Functional hyperemia
Pericytes
Inter-pericyte tunneling nanotubes
Blood-brain/retinal-barriers
ABSTRACT
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal
provision of oxygen and nutrients to sustain their activity. The timely regulation of blood ow is, therefore,
essential to supply ring neurons in active areas with the oxygen and glucose they need for energy. Many
glaucoma patients suffer from vascular decits including reduced blood ow, impaired autoregulation, neuro-
vascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated
by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller
cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the
ability of its members to regulate neurovascular interactions and how their function might be altered during
glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the
regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the
discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes,
which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary
network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysio-
logical implications, while highlighting opportunities to develop strategies for vascular protection and regen-
eration to improve functional outcomes in glaucoma.
1. Introduction
Glaucoma is a spectrum of neurodegenerative diseases characterized
by progressive optic nerve damage that can lead to irreversible blind-
ness. Due to its age dependence and scarcity of effective treatments, the
prevalence of glaucoma is predicted to grow over the next decades
(Tham et al., 2014). A common feature in the pathophysiology of all
glaucomas is the loss of retinal ganglion cells (RGCs), the neurons that
convey visual information from the retina to the brain. The special
cytoarchitecture of RGCs with elaborate dendrites in the retina, long
projecting axons, and synaptic terminals in brain centers, places a
considerable metabolic burden on these neurons (Almasieh et al., 2012;
Wareham et al., 2022; Tribble et al., 2023). RGCs have an exceedingly
high metabolism (Casson et al., 2020) and, as such, require a precise
regulation of blood supply to meet their oxygen and nutrient demand.
For decades, it has been thought that insufcient blood ow contributes
to RGC dysfunction and subsequent neurodegeneration in glaucoma
(Flammer et al., 1999). Prior to death, RGCs enter periods of dysfunction
(Calkins, 2012; Fry et al., 2018) associated with metabolic stress (Carelli
et al., 2009; Crish et al., 2010; Inman and Harun-Or-Rashid, 2017; Ito
and Di Polo, 2017; Osborne et al., 2016; Takihara et al., 2015; Williams
et al., 2017b; Kimball et al., 2018, Quintero et al., 2022), which can be
propelled by vascular decits. It is well known that many glaucoma
patients suffer from neurovascular dysfunction including decreased
blood ow in the retina and optic nerve, reduced vessel caliber, and
capillary defects (Flammer et al., 2002; Resch et al., 2009; Wareham and
Calkins, 2020; Shiga et al., 2016, 2018). Vascular autoregulation and
light-induced neurovascular responses are severely compromised in this
disease (Garh¨
ofer et al., 2004; Grunwald et al., 1984a; Gugleta et al.,
* Corresponding author. Department of Neuroscience Universit´
e de Montr´
eal Centre de recherche du centre hospitalier de lUniversit´
e de Montr´
eal (CRCHUM), 900
rue Saint-Denis, Montreal, QC, H2X 0A9, Canada.
E-mail address: adriana.di.polo@umontreal.ca (A. Di Polo).
Contents lists available at ScienceDirect
Progress in Retinal and Eye Research
journal homepage: www.elsevier.com/locate/preteyeres
https://doi.org/10.1016/j.preteyeres.2023.101217
Received 24 July 2023; Received in revised form 23 September 2023; Accepted 25 September 2023
Progress in Retinal and Eye Research 97 (2023) 101217
2
2012; Kiyota et al., 2017). Notwithstanding, the mechanisms underlying
vascular dysfunction in glaucoma and their impact on RGC and optic
nerve damage are poorly understood.
Vascular abnormalities are anticipated to promote neuronal
dysfunction, hence there has been a growing interest in exploring the
contribution of neurovascular decits to glaucomatous neuro-
degeneration. In this review, we aim to provide an updated account of
clinical observations on vascular impairment in glaucoma patients as
well as recent research on the role of neurovascular cells including
pericytes, which have been a recent focus of attention in this area.
Indeed, the discovery of tunneling nanotubes connecting pericytes in the
retina and their critical role in the regulation of coordinated neuro-
vascular interactions has had major implications in our understanding of
vascular decits during glaucomatous stress (Alarcon-Martinez et al.,
2020, 2022). We also discuss evidence of vascular barrier breakdown in
glaucoma and the potential cellular and molecular mechanisms involved
in these alterations.
2. Vascular impairment in glaucoma patients
2.1. Anatomy of the inner retina and optic nerve vascular systems
The delivery of oxygen and nutrients to the retina of higher mammals
is accomplished by two separate systems: the choroid and the retinal
vasculature, which derive from branches of the ophthalmic artery. The
choroidal circulation, which supplies the retinal pigment epithelium and
photoreceptors, is one of the most vascularized structures in mammals
required to meet the high metabolic demand of the outer retina (Bill,
1985; Kur et al., 2012). The inner retina, on the other hand, is nourished
by the retinal vasculature, which is supplied by the central retinal artery
(Singh and Dass, 1960a,b). The central retinal artery typically branches
into four radial arterioles extending outward from the optic nerve head
(ONH), each distributing blood to one quadrant of the retina. The retinal
arterioles give rise to three capillary beds: i) the supercial plexus that
supplies the ganglion cell and nerve ber layers, ii) the intermediate
plexus, which irrigates the inner plexiform layer, and iii) the deep plexus
Abbreviations
Ang2 Angiopoietin-2
ATP Adenosine Triphosphate
α
-SMA Alpha Smooth Muscle Actin
BRB Blood-Retina-Barrier
BBB Blood-Brain-Barrier
cGMP Cyclic Guanosine Monophosphate
CX43 Connexin-43
CSF1R Colony-Stimulating Factor 1 Receptor
CX3CR1 C-X3-C Motif Chemokine Receptor
EETs Epoxyeicosatrienoic Acids
EP
4
Prostaglandin E2 receptor 4
ERG Electroretinogram
ET
A
Endothelin Receptor A
ET
B
Endothelin Receptor B
ET-1 Endothelin-1
ET-2: Endothelin-2
ET-3: Endothelin-3
20-HETE 20-Hydroxy-Eicosatetraenoic Acid
ICWs Intercellular Ca
2+
Waves
IOP Intraocular Pressure
IP-TNTs Interpericyte Tunneling Nanotubes
IP3Rs Inositol 1,4,5-Trisphosphate Receptors
K
ATP
ATP-Sensitive K
+
channel
K
IR
Inward-Rectier K
+
channel
LSFG Laser Speckle Flowgraphy
MMP-9 Metalloproteinase-9
NO Nitric Oxide
NOS NO Synthase
NVC Neurovascular Coupling
NVU Neurovascular Unit
ONH Optic Nerve Head
PANX1 Purine Channel Pannexin 1
PDGF-B: Platelet-Derived Growth Factor B
PDGFR-β: PDGF receptor-β
PECAM-1 Platelet Endothelial Cell Adhesion Molecule-1
PGE
2
Prostaglandin E2
P2RY12 P2Y12 Purinergic Receptor
RGCs Retinal Ganglion Cells
ROS Reactive Oxygen Species
TGF-β: Transforming Growth Factor-β
TRP Transient Potential Channels
VE-cadherin Vascular Endothelial-Cadherin
VEGF-A: Vascular Endothelial Growth Factor-A
VDCC Voltage-Dependent Ca
2+
channel
ZO-1 Zona Occludens-1
Fig. 1. Retina and optic nerve head vascular organization. (A) Schematic representation of the distinct vascular plexuses in the retina: supercial, intermediate, and
deep. (B) Organization of the vasculature in the optic nerve head region. CRA: central retinal artery; CRV: central retinal vein; PCA: posterior ciliary artery. Created
with BioRender.com.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
3
that nourishes the outer plexiform layer (Newman, 2013; Korneld and
Newman, 2014) (Fig. 1A). In the human retina, the radial peripapillary
capillary plexus comprises a fourth, anterior capillary bed specialized to
serve the thickest portion of the retinal nerve ber layer (Henkind, 1967;
Scoles et al., 2009). Recent evidence from analysis of optical coherence
tomography angiography scans indicates that the thickest part of the
nasal parafoveal rim tissue is also endowed with a fourth, anterior layer
of capillaries in addition to the three widely recognized layers described
above (Hirano et al., 2018). The blood is returned to the central retinal
vein in the ONH and ultimately drained into the cavernous sinus (Harris
and Rhoton, 1976). Unlike the choroid, which is regulated by both
sympathetic and parasympathetic nerves (Alm, 1977; Kawarai and Koss,
1998; Nilsson, 1996), the retinal vasculature lacks autonomic innerva-
tion and is controlled by mechanisms whose actions are collectively
known as autoregulation (Laties, 1967; Ye et al., 1990), which is dis-
cussed below in more detail.
The ONH is considered to be critically important in glaucoma
pathogenesis, perhaps even the primary site of injury to RGC axons
(Minckler and Bunt, 1977; Quigley and Addicks, 1981; Nickells et al.,
2012; Wilkison et al., 2021). Anatomically, the ONH comprises the
prelaminar, laminar, and retrolaminar regions (Hayreh, 1997; Mack-
enzie and Ciof, 2008). Blood supply to the ONH is highly anastomotic,
deriving in large part from the short posterior ciliary arteries, which
penetrate the peripapillary sclera to feed a dense network of ramied
capillaries often via a complete or partial arteriolar circle of Zinn-Haller
including those within the lamina cribrosa (Ciof Ga, 1996). The su-
percial nerve ber layer receives blood from the central retinal artery
and the retrolaminar region also from pial arterioles (Fig. 1B). The
lamina cribrosa, a trabeculated structure of connective tissue that pro-
vides structural and metabolic support to RGC axons, is supplied by
branches from the short posterior ciliary arteries through the circle of
Zinn-Haller. Vessels in this area are exposed to a highly dynamic
biomechanical environment of varying load and strain, and a
trans-laminar pressure gradient established by the difference between
the intraocular pressure (IOP) and the cerebrospinal uid pressure
manifest in the retrobulbar optic nerve via the subarachnoid space of its
sheath (Jonas et al., 2015). Indeed, biomechanical strains and stress in
this area have been proposed to contribute to both direct and indirect
mechanisms of axon injury, including vascular decits during glau-
comatous disease pathogenesis (Sigal and Ethier, 2009; Burgoyne, 2011;
Downs, 2015).
2.2. Vessel caliber and basal blood ow alterations
It is well established that glaucoma patients suffer from vascular
decits that manifest as reduced vessel caliber, decreased blood ow in
the retina and ONH, and capillary defects. This topic has been previously
covered in excellent reviews (Flammer et al., 2002; Newman et al.,
2018; Wareham and Calkins, 2020), therefore only a summary of recent
ndings will be discussed in this section. Advances in blood ow mea-
surements and imaging technologies, including optical coherence to-
mography angiography, have provided the ability to quantify
microvascular changes in glaucoma patients alongside structural alter-
ations (Rao et al., 2020; Shin et al., 2022; Shiga et al., 2023). A number
of studies have demonstrated reduced vessel density in the ONH and in
the peripapillary region in primary open-angle glaucoma (Jia et al.,
2012, 2014; Liu et al., 2015; Yarmohammadi et al., 2016) as well as
low-tension glaucoma patients (Maekawa et al., 2014; Yabana et al.,
2017). Vessel density in the macular region and deep layers of the
parapapillary choroid were shown to be decreased in glaucomatous eyes
(Suh et al., 2016; Lee et al., 2017b,c; Rao et al., 2016, 2017; Wan et al.,
2018; Moghimi et al., 2018). At the level of retinal capillaries, neuro-
vascular coupling has been proposed to be vascular plexus-dependent
(Hormel et al., 2021). Recent imaging data using optical coherence to-
mography show that the capillary density in the supercial vascular
complex, which comprises the nerve ber layer plexus and the ganglion
cell layer plexus, is reduced in glaucoma patients (Liu et al., 2019).
Plexus-specic responses may reect differences in the metabolic de-
mand of cells, intrinsic vascular properties, and levels of vasoactive
metabolites at each location. However, the extent to which
plexus-specic light-evoked capillary responses are involved in glau-
coma pathogenesis remains an open question.
Blood ow measurements provide a readout of the functional status
of the vascular system. Quantitative studies in open-angle and low-
tension glaucoma patients demonstrated signicant blood ow alter-
ations. For example, early-stage open-angle glaucoma patients demon-
strated a reduction of retinal and ONH microvascular content relative to
normal controls (Jia et al., 2012; Liu et al., 2015; Kiyota et al., 2021). In
low-tension glaucoma patients, a reduction in ONH blood ow observed
at the earliest stages of the pathology was found to be a risk factor for
disease progression (Shiga et al., 2016, 2018). Although robust longi-
tudinal human studies are still lacking, the overall consensus from
cross-sectional studies is that there is a strong correlation between the
loss of microvasculature and blood ow and the severity of glaucoma
(Liu et al., 2015; Takusagawa et al., 2017; Shin et al., 2017; Mansoori
et al., 2017; Geyman et al., 2017; Shiga et al., 2016, 2018; Rao et al.,
2020; Kiyota et al., 2021). While basal blood ow alterations are
important, the next sections argue that decits in the dynamic ability of
the system to regulate blood supply in response to neuronal activity
plays an even more critical role in functional visual decits associated
with glaucoma.
2.3. Vascular autoregulation decits
In healthy individuals, vascular beds in certain tissues of the body
exhibit an intrinsic ability to maintain constant blood ow over a large
range of perfusion pressures and varying metabolic demand, a process
known as autoregulation. In the retina and ONH, vascular autor-
egulation refers to the capacity of these tissues to sustain a stable blood
supply despite changes in ocular perfusion pressure (Prada et al., 2016).
The ocular perfusion pressure is the force driving blood through the
intraocular vasculature, estimated as the difference between arterial
blood pressure and IOP under the assumption that the intraocular
venous pressure is close to the IOP (Attariwala et al., 1994; Costa et al.,
2014; Glucksberg and Dunn, 1993; Guidoboni et al., 2014). In glaucoma
patients, the central retinal vein pressure can be higher than the IOP,
which may lead to lower perfusion pressure in the retina and ONH, ul-
timately contributing to disease worsening (Pillunat et al., 2014). The
autoregulation capacity of the system is conventionally assessed by
repeated blood ow measurements, obtained before and after an in-
crease or decrease in ocular perfusion pressure, which reect the slower
time course of effects known as static autoregulation (Prada et al.,
2016). This component is part of the myogenic response, which refers to
the intrinsic ability of vessels to adjust vascular tone in response to
changes in transmural pressure (Hayreh, 2001).
The retina and ONH circulation exhibit considerable autoregulation
capacity in response to changes in ocular perfusion pressure (Geijer and
Bill, 1979; Riva et al., 1997; Sossi and Anderson, 1983; Pillunat et al.,
1997; Schmidl et al., 2011). Low ocular perfusion pressure - due to
increased IOP and/or low blood pressure - augment the risk of devel-
oping glaucoma as well as disease progression (Bonomi et al., 2000;
Leske et al., 2007, 2008; Quigley et al., 2001; Leske, 2009; Choi et al.,
2007; Sung et al., 2009). Indeed, glaucoma patients display impaired
blood ow autoregulation when the homeostatic limits are challenged
by changes in perfusion pressure, posture, hypercapnia, hyperoxia, or
cold temperatures (Grunwald et al., 1984a; Nagel et al., 2001; Gherghel
et al., 2004; Sines et al., 2007; Feke and Pasquale, 2008; Venkataraman
et al., 2010; Cherecheanu et al., 2012; Prada et al., 2016). For example,
systemic hyperoxia induced by breathing pure oxygen results in marked
vasoconstriction of retinal arterioles and ONH capillaries in healthy
human subjects (Eperon et al., 1975; Riva et al., 1983, 1986b; Stefansson
et al., 1988; Hickam and Frayser, 1966; Jean-Louis et al., 2005; Fallon
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
4
et al., 1985; Kiss et al., 2002; Shiga et al., 2015). A cross-sectional study
revealed that patients with open-angle glaucoma display weaker vaso-
active responses to systemic hyperoxia than aged-matched controls
(Kiyota et al., 2017). Furthermore, weaker ONH vasoreactivity to sys-
temic hyperoxia was associated with the progression of visual eld de-
fects in patients with open-angle glaucoma (Kiyota et al., 2020).
Dynamic autoregulation refers to the actions causing rapid and
transient vascular changes in response to sudden shifts such as a pressure
step challenge (e.g., a rapid increase of IOP or decrease of blood pres-
sure), reecting the processes that precede the equilibrated steady state
or static autoregulation discussed above (Liang et al., 2010; Prada et al.,
2016). Analysis of dynamic autoregulation involves high temporal res-
olution blood ow measurements, made during the initial phases of
pressure changes, effectively revealing autoregulatory dysfunction
(Noack et al., 2007; van Beek et al., 2008; Liang et al., 2010). Studies of
cerebral blood ow indicate that dynamic autoregulation is a better
indicator of hemodynamic decits than static autoregulation because it
correlates more closely with neuronal activity (Panerai, 1998; Rose-
ngarten et al., 2007). Of interest, elegant studies in non-human primates
subjected to chronic IOP elevation demonstrated ONH blood ow dy-
namic autoregulation dysfunction (Wang et al., 2014b), whereas static
autoregulation remained unchanged (Wang et al., 2014a). Recent work
conducted in a clinical setting with healthy human volunteers also
leveraged the high temporal resolution of the laser speckle owgraphy
(LSFG) imaging technique to evaluate changes in blood ow and pa-
rameters of pulsatility (over the cardiac cycle) within capillaries of the
ONH and the major retinal vessels following acute perturbations of IOP
by ophthalmodynamometry (Iwase et al., 2021). The results demon-
strated that acute IOP elevation caused rapid reduction of blood ow
and higher resistance to blood ow within ONH capillaries, as indicated
by a host of pulsatility parameters (Iwase et al., 2021). Similarly, LSFG
pulsatility parameters, indicative of higher resistance to ow, were
found to be elevated in older age (Luft et al., 2016), an important risk
factor for glaucoma and its progression. LSFG pulsatility was also pre-
dictive of the rate of subsequent progression of visual eld loss in
glaucoma suspects (Gardiner et al., 2023) and was found to be increased
in early stage normotensive glaucoma (Shiga et al., 2013). This pattern
of results parallels altered dynamic autoregulation observed in animal
models (Wang et al., 2014b). It is possible that increased resistance is
caused by mechanically ‘stiffer vasculature, loss of capillaries from
normal beds and/or via functional effects such as aberrantly constricted
pericytes (see below). Collectively, these data suggest that blunted ONH
and retinal vasoreactivity contribute to glaucoma pathophysiology and
that dynamic autoregulation is a useful outcome measure to identify
vascular dysfunction in glaucoma patients when conventional ap-
proaches are insufcient.
2.4. Neurovascular dysfunction in glaucoma subjects
Discovered almost 150 years ago, neurovascular coupling (NVC) is a
physiological mechanism that adapts blood ow in accordance with
local neuronal activity, thus bringing additional oxygen and nutrients to
active neurons (Mosso, 1880; Roy and Sherrington, 1890; Friedland and
Iadecola, 1991). This process, also known as functional hyperemia, is
particularly active in the vasculature of the retina and optic nerve in
response to neuronal activation induced by light stimuli (Bill, 1975; Riva
et al., 2005; Srienc et al., 2010; Yu et al., 2019; Wareham and Calkins,
2020). In humans, the NVC response has been studied in retinal and
ONH vessels using ickering light stimulation, which triggers vasodi-
lation and increased blood ow under physiological conditions (Riva
et al., 1986a, 1997, 2005; Polak et al., 2002; Garh¨
ofer et al., 2003,
2005). Another study used adaptive optics to enable visualization of
individual retinal capillaries and quantication of their response to focal
light stimulation in healthy human volunteers, and demonstrated evi-
dence of neurovascular coupling at the level of capillaries, as well as
complex response patterns (inhomogeneity of dilations and
constrictions) suggestive of blood ow redistribution according to local
metabolic demands (Duan et al., 2016). Patients with open-angle glau-
coma present a signicant reduction in icker-evoked dilation of retinal
vessels at the early stages of the disease (Grunwald et al., 1984b;
Garh¨
ofer et al., 2004; Riva et al., 2005; Gugleta et al., 2012). Retinal
vascular responses to icker light also decrease with aging (Gugleta
et al., 2013) and vasospastic propensity (Gugleta et al., 2006), two major
risk factors for glaucoma development (Guedes et al., 2011; Flammer
et al., 2001). Capillaries in the ONH also possess NVC capacity in
response to icker light stimulation (Riva et al., 2005; Gugleta et al.,
2007). Changes in ONH blood ow following luminance icker in
glaucoma patients, including individuals in the initial phases of the
disease are decreased relative to healthy controls (Riva et al., 2004).
Together, these data support the involvement of NVC dysfunction at the
early stages of glaucomatous damage. The next sections discuss the
cellular and molecular mechanisms that regulate NVC and how decits
in these processes can have critical repercussions for glaucoma
pathogenesis.
3. The neurovascular unit: at the heart of stress-related changes
in glaucoma
The function of the retina and optic nerve is not mediated by neurons
alone and, instead, requires multiple cells that communicate with each
other to fulll the metabolic requirement of these tissues through co-
ordinated neurovascular responses. This community of cells is known as
the neurovascular unit (NVU), a concept rst introduced in 2001 by the
Stroke Progress Review Group meeting of the National Institute of
Neurological Disorders and Stroke of the National Institutes of Health to
acknowledge the symbiotic relationship between neurons and sur-
rounding cells (Iadecola, 2017). The NVU in the retina and optic nerve is
composed of neurons, endothelial cells, pericytes, smooth muscle cells,
and glial cells including astrocytes, Müller cells, and microglia (Fig. 2).
The complex signaling between the multiple cell types of the NVU en-
gages distinct effector systems that coordinate neurovascular responses
and maintain the integrity of vascular barriers (Newman, 2013; Iade-
cola, 2017; Sweeney et al., 2019; Pfeiffer et al., 2021; Alarcon-Martinez
et al., 2021; Kugler et al., 2021; OLeary and Campbell, 2023).
Increased blood ow occurring through the actions of the NVU is also
believed to be important to clear the brain and retina of potentially toxic
by-products of neuronal activity such as lactate and carbon dioxide as
well as for temperature regulation (Zhu et al., 2006; Tarasoff-Conway
et al., 2015). In addition, it is now clear that the NVU interacts bidi-
rectionally with the systemic metabolism, peripheral immune system,
and gut microbiota (Tiedt et al., 2022) expanding its implications in
health and disease. Consistent with this, dysfunction of NVU compo-
nents leading to abnormal vascular responses occurs in several pathol-
ogies including diabetes, Alzheimers disease, and other conditions
(Garh¨
ofer et al., 2020; Li et al., 2023). For example, in diabetic reti-
nopathy, pericyte loss and endothelial cell dysfunction coupled with
neuronal and glia alterations are thought to impair functional hyper-
emia (Mishra and Newman, 2010; Garh¨
ofer et al., 2020), highlighting
the involvement of multiple NVU cellular networks in the disease pro-
cess. In addition, pericyte and endothelial cell impairment lead to
capillary constriction during myocardial infarction, stroke, epilepsy, or
COVID-19 infection (Hall et al., 2014; OFarrell et al., 2017; Leal--
Campanario et al., 2017; Hirunpattarasilp et al., 2023). Endothelial
dysfunction has also been observed in the systemic vasculature, partic-
ularly in patients with low-tension glaucoma (Henry et al., 1999; Su
et al., 2006), suggesting that multi-systemic vascular risk factors may
inuence local ocular NVC breakdown in glaucomatous eyes. Collec-
tively, these studies support the notion that while RGC activity initiates
the process of neurotransmission from retina to brain, the imple-
mentation of a full hemodynamic response requires nely tuned in-
teractions with other NVU cells. In the next sections, we will review the
importance of the primary NVU cellular components in the context of
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
5
neurovascular regulation and the implications for glaucoma.
3.1. RGCs as orchestra conductors of hemodynamic changes
Neurons are traditionally considered the initiators of the local neu-
rovascular response by signaling to blood vessels, directly or indirectly,
thus triggering coordinated hemodynamic changes (Iadecola, 2017). An
orchestrated neurovascular response is achieved by two means: i)
metabolic negative feedback, and ii) a feedforward mechanism.
Increased energy consumption induced by neuronal activity lowers ox-
ygen and/or glucose leading to the accumulation of metabolic byprod-
ucts, which act as signals for blood vessel dilation required to provide
more energy (metabolism-dependent feedback) (Attwell et al., 2010;
Iadecola, 2017; Pfeiffer et al., 2021). At the same time, glutamate
released by neuronal activity acts on glutamate receptors leading to
calcium (Ca
2+
)-dependent release of vasoactive factors that drive the
initial feedforward mechanism (metabolism independent) of the local
blood ow response (Iadecola, 2017). The metabolism-dependent
feedback and feedforward mechanisms are not mutually exclusive
(Iadecola, 2017), and both are likely to contribute to the vascular energy
supply that supports RGCs depending on the timing, intensity, and
duration of their activation.
The metabolic negative feedback is triggered by a sudden increase in
ATP consumption at the onset of neurotransmission lowering tissue
oxygenation and glucose, while increasing carbon dioxide and meta-
bolic byproducts, thus blood ow is matched with the metabolic need of
active neurons (Iadecola, 2017). In support of this, brain blood ow was
shown to increase in response to a transient decrease in tissue oxygen-
ation (Lecoq et al., 2011; Parpaleix et al., 2013). Studies in the lateral
geniculate nucleus and visual cortex of cats found a reduction in oxygen
and glucose, preceding blood ow increase, at the onset of light-evoked
neural activity (Freeman and Li, 2016). Of interest, analysis of the ce-
rebral microvasculature showed that transient dips in tissue oxygen
tension at the beginning of neuronal activation increased the deform-
ability of red blood cells improving blood ow velocity and capillary
hyperemia (Wei et al., 2016a). In the retina, optic nerve, and brain, the
elevation of the carbon dioxide partial pressure was shown to induce a
global blood ow increase (Harris et al., 1998; Hoiland et al., 2019). In
addition, active neurons release two potent vasodilator byproducts,
adenosine and lactate (Ko et al., 1990; Ido et al., 2001), which can
contribute to NVC by acting as mediators of the metabolic negative
feedback (Attwell et al., 2010; Iadecola, 2017).
Metabolic stress and energy decits have been shown to contribute
to RGC damage in glaucoma (Inman and Harun-Or-Rashid, 2017; Ito and
Di Polo, 2017; Morquette and Di Polo, 2008; Williams et al., 2017a;
Williams et al., 2017b; Belforte et al., 2021, Quintero et al., 2022).
Consistent with this, hyperactivation of the energy sensor adenosine
monophosphate kinase, which is triggered by low intracellular ATP, was
found in RGCs from mice subjected to ocular hypertension and primary
open-angle glaucoma patients (Belforte et al., 2021). While direct proof
of ATP shortage in living RGCs had been lacking, a recent longitudinal
study using two-photon microscopy live imaging demonstrated a sub-
stantial decrease in ATP levels in RGC soma and axons subjected to
ocular hypertensive stress (Quintero et al., 2022). A drop in RGC energy
reserves in glaucoma may compromise the ability of these neurons to
orchestrate NVC regulation restricting the timely delivery of oxygen and
glucose which will, ultimately, lead to neurodegeneration. However, it
is also possible that NVC failure may reduce the energy available to
RGCs (see section 4.4). Recent studies demonstrated that adenosine and
lactate can promote RGC survival (Vohra et al., 2019; Agarwal and
Agarwal, 2021; Boia et al., 2020). Furthermore, it is still unknown
whether endogenous lactate or adenosine, locally produced by RGCs or
surrounding cells, restore NVC responses enhancing neuronal viability
and function during glaucomatous damage. While intracellular ATP
decreases in RGCs subjected to high intraocular pressure (Quintero
et al., 2022), high extracellular ATP has been observed in eyes with
glaucoma (Li et al., 2011; Lu et al., 2015). Excessive extracellular ATP,
released from damaged RGCs and neighboring glial cells, can bind to
P2X7 purinergic receptors on RGCs and microglia (Campagno et al.,
2021; Ishii et al., 2003; Puthussery and Fletcher, 2004) triggering
neuronal death (Hu et al., 2010). Given that excess extracellular ATP is
involved in pericyte-dependent capillary contraction via purinergic
signaling activation in brain diseases such as Alzheimers disease
(Hørlyck et al., 2021), abnormal extracellular ATP levels associated with
glaucomatous stress may harm microvasculature as well. Additional
studies are needed to fully understand the link between RGC metabolic
stress and NVC impairment in glaucoma.
In the brain, feedforward regulation is primarily driven by the
release of potent vasodilators, notably nitric oxide (NO) and prosta-
glandins (Yang et al., 2003; Attwell et al., 2010; Lecrux and Hamel,
2016). Neuronal activity-evoked ionic changes such as an increase in K
+
can also mediate NVC (Longden et al., 2016) and will be discussed in the
context of the endothelial cell responses in the next section. Early studies
showed that NO increased following light icker stimulation in the
retina and ONH (Donati et al., 1995; Buerk and Riva, 2002) lending
support to a role for NO in neurovascular changes at these locations.
Consistent with these ndings, studies in cats as well as humans showed
that systemic administration of non-selective NOS inhibitors reduced
icker-evoked increases in blood ow (Kondo et al., 1997; Dorner et al.,
2003). More recent studies suggest that NO does not mediate NVC in the
retina (Metea and Newman, 2006a; Nippert and Newman, 2020) but
rather serves as a vasomodulator, similar to its role in the cerebral cortex
(Lindauer et al., 1999). For example, when NO levels were increased or
decreased in the medium of acutely isolated whole-mounted retinas, it
Fig. 2. Cellular components of the neurovascular unit (NVU) in the retina and optic nerve. (A) Schematic representation of the cells that form the neurovascular unit
in the retina, which include retinal ganglion cells (RGC), astrocytes, endothelial cells, pericytes, Müller cells, and microglia. (B) The neurovascular unit in the optic
nerve includes RGC axons, both the unmyelinated and myelinated portions, astrocytes, pericytes, endothelial cells, microglia, and oligodendrocytes, the latter
depending on whether the axon segment is myelinated or not. Created with BioRender.com.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
6
promoted light-evoked arteriole vasoconstriction or vasodilation,
respectively (Metea and Newman, 2006a), possibly by modulating the
synthesis of arachidonic acid derivatives (Attwell et al., 2010). NO can
stimulate the production of cyclic guanosine monophosphate (cGMP),
which inhibits Ca
2+
entry in pericytes hence reducing their contractility
(Sakagami et al., 2001). Thus, NO can function as a mediator of NVC and
has been implicated in glaucomatous neurodegeneration (Haider et al.,
2022).
In models of diabetic retinopathy, loss of NVC has been attributed to
an increase in NO, which blocked the production of vasodilation agents
derived from Müller cells (see Section 3.4.1.). In support of this,
inducible NO synthase (NOS) inhibitors restored functional hyperemia
in retinal explants (Mishra and Newman, 2010; Mishra et al., 2011). The
role of NOS in glaucomatous optic neuropathy has been controversial.
Early studies showed increased expression of inducible NOS in the ONH
of open-angle glaucoma patients (Neufeld et al., 1997), and NOS inhi-
bition with aminoguanidine provided RGC neuroprotection in a rat
glaucoma model (Neufeld et al., 1999). However, subsequent studies
using several independent quantitative methods and robust glaucoma
models failed to identify signicant changes in NOS expression in the
retina, ONH, and optic nerve; nor was neuroprotection conferred by
genetic deletion or pharmacological inhibition of NOS (Pang et al.,
2005; Libby et al., 2007). These ndings, together with studies showing
that NO inhibition only partly accounts for NVC (Hosford and Gourine,
2019), suggest that other cellular and molecular pathways of neuro-
vascular regulation are at play and will be discussed in the following
sections.
3.2. Endothelial cells set the tone
The wall of blood vessels is lined by a thin monolayer of endothelial
cells, known as the vascular endothelium, which form an adaptive and
dynamic interface between the blood circulation and the tissue envi-
ronment (Krüger-Genge et al., 2019). Endothelial cells are subjected to
hemodynamic changes and receive and send signals to regulate the
myogenic or vessel tone, vascular permeability, and immune responses
(Loh et al., 2018; OLeary and Campbell, 2023). Cells in the vascular
endothelium are electrically coupled through gap junctions, which
allow the transfer of electrical signals from capillaries to arterioles and
venules (Zhang et al., 2011; Longden et al., 2017). Being in close contact
with mural cells, namely smooth muscle cells and pericytes, endothelial
cells are active players within the NVU (Dalkara and Alarcon-Martinez,
2015). The myogenic response is determined by the balance between
vasodilation and vasoconstriction signals that act on receptors expressed
by endothelial and mural cells (Davis and Hill, 1999; Koide et al., 2018).
Vasodilation is mediated by endothelium-derived relaxing factors
including endothelial NO (Zhao et al., 2015a; Loh et al., 2018), pros-
tacyclin (Ingerman-Wojenski et al., 1981, Oudemans-Van Straaten et al.,
2010), endothelium-derived hyperpolarizing factor (F´
el´
etou and Van-
houtte, 2006), and hydrogen sulde (Zhao and Wang, 2002), while
vasoconstriction is regulated by other mediators such as angiotensin II,
generated from the endothelium-located angiotensin-converting
enzyme that converts angiotensin I into angiotensin II (Benigni et al.,
2010; Vanlandewijck et al., 2018), thromboxane A
2
(Ramadan et al.,
1990; Ingerman-Wojenski et al., 1981), endothelin (Vanhoutte and
Tang, 2008; Krüger-Genge et al., 2019), prostaglandin H
2
(Davidge
et al., 1995), and reactive oxygen species (ROS) (Fan et al., 2014).
Among these, endothelin deserves special attention based on accumu-
lating evidence suggesting a role in glaucoma (Chauhan, 2008; Prasanna
et al., 2011).
Originally isolated from the supernatant of cultured porcine endo-
thelial cells, endothelin-1 (ET-1) is a potent vasoconstrictor with long
lasting effects (Yanagisawa et al., 1988). Two additional family mem-
bers, endothelin-2 (ET-2) and endothelin-3 (ET-3), were identied in
humans (Inoue et al., 1989). Endothelins bind to and activate the
G-protein-coupled receptors ET
A
and ET
B
(Arai et al., 1990; Sakurai
et al., 1990) with ET
A
showing higher afnity for ET-1 and ET-2 than
ET-3 (Arai et al., 1990). ET-1 is presumed to promote vasoconstriction
by acting on ET
A
receptors expressed in mural cells, notably pericytes
(Howell et al., 2014; Vanlandewijck et al., 2018). Endothelins and their
receptors are also expressed by other cells in the retina and optic nerve,
including RGCs and glia (MacCumber and DAnna, 1994; Rattner and
Nathans, 2005; Minton et al., 2012; Rattner et al., 2013; Howell et al.,
2014; Patel et al., 2014). ET-1 levels increase in the aqueous humor of
primary open-angle and exfoliation glaucoma patients (Tezel et al.,
1997; Koukoula et al., 2018). Intravitreal injection or overexpression of
endothelins promote RGC death, whereas inhibition of endothelin
signaling is neuroprotective (Orgül et al., 1996; Chauhan et al., 2004;
Sasaoka et al., 2006; Lau et al., 2006; Howell et al., 2011, 2012; Mi et al.,
2012; Blanco et al., 2017; Marola et al., 2020, 2022). However, the
precise site of action of endothelins and how they promote RGC death is
still poorly understood. Of interest, a recent study demonstrated that
cell-specic conditional deletion of both ET
A
and ET
B
from RGCs and
macroglia did not prevent ET-1-induced RGC loss (Marola et al., 2022).
In contrast, deletion of ET
A
from vascular mural cells was sufcient to
block vasoconstriction and RGC death caused by ET-1 exposure (Marola
et al., 2022). Therefore, ET-1, independently of where it is produced,
does not act directly on RGCs or glia but rather uses vascular mural cells
to drive RGC death. These ndings suggest that ET
A
-mediated capillary
vasoconstriction is a critical event leading to RGC loss.
Although endothelial cells have been traditionally considered to play
a limited role in NVC, recent elegant studies have challenged this idea by
reporting that endothelial cells express receptors and channels that
transduce neuronal activity into a vascular response (Longden et al.,
2016, 2017, 2021; Harraz et al., 2018a,b; Zhang et al., 2021; Thakore
et al., 2021; Sancho et al., 2022b). In brain vessels, potassium (K
+
) has
been proposed to be an important mediator of NVC acting on the strong
inward-rectier K
+
(K
IR
) channels in the vascular wall, which serve as
sensors of external K
+
(Longden and Nelson, 2015). Neuronal activity
increases extracellular K
+
and activation of K
IR
channels, predominantly
the K
IR
2.1 subtype expressed in both endothelial and mural cells
(Bradley et al., 1999; He et al., 2018), leading to hyperpolarization and
pronounced vasodilation (Longden and Nelson, 2015; Longden et al.,
2016). This capillary response propagates retrogradely across endothe-
lial cells through gap junctions, dilating upstream arterioles and
increasing blood ow along the entire vascular tree (Longden et al.,
2017; Moshkforoush et al., 2020; Zhang et al., 2021). K
IR
2.1 dysfunction
has been associated with a number of pathologies including chronic
stress, cerebral small vessel disease, and traumatic brain injury (Long-
den et al., 2014; Dabertrand et al., 2021; Weir and Longden, 2021). Low
levels of phosphatidylinositol 4,5-bisphosphate (PIP
2
), an intracellular
phospholipid that is essential for K
IR
2.1 channel function, have been
reported in a model of small vessel disease (Harraz et al., 2018a,b), and
PIP
2
administration was sufcient to restore K
IR
2.1 channel signaling
and cerebral blood ow in response to neuronal activation (Dabertrand
et al., 2021). K
IR
2.1 channels are expressed in the retina and optic nerve
(Brasko and Butt, 2018; Klose et al., 2021), however, studies on the role
of Kir2.1 signaling in endothelial cell hyperpolarization in the context of
glaucoma and their implication in NVC dysfunction are still lacking.
3.3. Pericytes: location, location, location
Described over 150 years ago, pericytes are cells that wrap tightly
around the surface of capillaries in a bump-on-a-log fashion (Eberth,
1871; Rouget, 1873; Attwell et al., 2016). Pericytes are a heterogeneous
population of mesenchymal cells that can have different morphologies
and functions depending on their location (Bohannon et al., 2021;
Garrison et al., 2023). In the brain, two main structural and functional
pericyte types have been described downstream of the penetrating
arteriole: ensheathing pericytes on the arteriole-capillary transition, and
thin-strand capillary pericytes located on capillaries (Hartmann et al.,
2022). Pericytes at the arteriole-capillary transition, also known as
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
7
ensheathing pericytes, display a mesh-like pattern of short-range pro-
cesses wrapped around the vessels (Grant et al., 2019; Alarcon-Martinez
et al., 2021; Longden et al., 2023). In contrast, capillary pericytes extend
thin processes along the capillary surface that can reach hundreds of
microns in length (Berthiaume et al., 2018; Alarcon-Martinez et al.,
2021). The human retinal vasculature is composed primarily of capil-
laries and ~95% of those are wrapped by pericytes (Chan-Ling et al.,
2011; Trost et al., 2019). Therefore, here we will use the term ‘pericyte
to refer to capillary pericytes, which will be the focus of this and sub-
sequent sections (see sections 4 and 5.2).
Capillary pericytes are embedded within the basement membrane of
the capillary wall with their soma and processes in intimate contact with
adjacent endothelial cells (Mandarino et al., 1993). Indeed, pericyte thin
processes are connected with more than one endothelial cell via gap
junctions as well as peg-and-socket interdigitations, where pericytes or
endothelial cells send a projection to make direct contact with the
adjacent cell (Ornelas et al., 2021). These contact points are likely to be
sites of gap junction communication and cross-talk between pericytes
and endothelial cells, where key signaling events take place including
increases in local Ca
2+
and second messengers (Cuevas et al., 1984;
Armulik et al., 2005). In whole retinas, functional connectivity maps
show that pericytes establish gap junction connections primarily with
neighboring pericytes and endothelial cells, and much less with arteri-
olar smooth muscle cells (Kovacs-Oller et al., 2020). Moreover, pericytes
can interact with surrounding neurons and glia (Allsopp and Gamble,
1979; Hogan and Feeney, 1963; Maynard et al., 1957; Murakami et al.,
1979; Sims, 1986; Ushiwata and Ushiki, 1990). For example, in human
retinas, tight contacts between glial cells and pericytes have been re-
ported in addition to fenestrations in the basement membrane of peri-
cytes at the contact site with Müller glia (Hogan and Feeney, 1963). The
central location of pericytes, which readily allows them to respond to
vasodynamic factors derived from neurons and other cells in the NVU,
makes them ideal candidates to regulate capillary blood ow and NVC
(Fig. 2). Consistent with this, single-cell transcriptomic studies demon-
strate that capillary pericytes express multiple ion channels and G
protein-coupled receptors for vasoactive mediators (Hariharan et al.,
2020; Vanlandewijck et al., 2018). Therefore, pericytes are ideally
positioned to play a critical role in regulating blood ow to meet the
energy requirements of RGCs.
Until recently, and despite their strategic location and vasodynamic
responses, pericytes were not considered serious contenders to regulate
blood ow in vivo, a process that was attributed solely to arteriolar
smooth muscle cells (Hill et al., 2015; Pfeiffer et al., 2021; Hartmann
et al., 2022). The controversy to resolve this issue stemmed from mul-
tiple challenges including a lack of tools to unequivocally identify and
target capillary pericytes, low spatial resolution to visualize small
changes in capillaries, and failure to detect components of the con-
tractile machinery required to constrict microvessels. However, recent
advances in the eld have yielded new data strongly supporting that
pericytes are contractile and that they play key roles in the regulation of
microcirculatory blood ow in the brain and retina (Peppiatt et al.,
2006; Hall et al., 2014; Kisler et al., 2017; Alarcon-Martinez et al., 2018,
2019, 2020, 2022; Vanlandewijck et al., 2018; Nortley et al., 2019;
Gonzales et al., 2020; Hartmann et al., 2021; Longden et al., 2023). For
example, histological and single-cell transcriptomic studies show that
pericytes express genes involved in actomyosin contraction, such as
lamentous actin (F-actin), myosin, regulators of myosin phosphoryla-
tion, and L-type voltage gated Ca
2+
channels (Vanlandewijck et al.,
2018; Kureli et al., 2020). However, expression of smooth muscle actin
organized in alpha conformation (
α
-SMA), required to bind myosin
proteins, was reported to be heterogeneous among pericyte populations
and was often found to be present at low or undetectable levels (Nehls
and Drenckhahn, 1991; Bandopadhyay et al., 2001). A recent study in
the retina addressed this issue by showing that when F-actin depoly-
merization is prevented using stabilizing agents or by snap xation,
α
-SMA is reliably detected in a large population of pericytes in all retinal
plexuses (Alarcon-Martinez et al., 2018). Furthermore, when
α
-SMA
expression was blocked, the ability of retinal pericytes to constrict
capillaries was markedly reduced (Alarcon-Martinez et al., 2018, 2019).
Recent functional studies have unequivocally established a critical role
for pericytes in the regulation of blood ow and NVC in the retina and
optic nerve, both in physiological and pathological conditions, including
glaucomatous damage (Alarcon-Martinez et al., 2020, 2022) and will be
discussed in Section 4.
3.4. Glia-derived vasoactive molecules
Once believed to play only supportive roles for neurons, glial cells
are now recognized to have a major inuence on most physiological
aspects of brain function including neurotransmission, energy homeo-
stasis, and blood ow (Kugler et al., 2021; Nampoothiri et al., 2022).
Glial cells in the mammalian retina and optic nerve, comprising astro-
cytes, Müller cells, microglia, and oligodendrocytes, play multiple roles
in the regulation of visual function (Yazdankhah et al., 2021; Benfey
et al., 2022). These cells are equipped with the molecular machinery to
sense and decode neuronal activity (Benfey et al., 2022). By responding
to multiple neurotransmitters, glial cells undergo coordinated changes
in intracellular Ca
2+
that promote the release of potent glia-derived
vasoactive agents (Newman, 2015). The physical proximity of glial
cells with pericytes and capillaries as well as their ability to respond to
neurons, make them ideal hubs for the tight control of blood ow
required to support optimal neural circuit function.
3.4.1. Signals from Müller cells and astrocytes
Müller cells are among the rst responders following retina or optic
nerve injury and can have neuroprotective as well as detrimental effects
on RGCs depending on the stress stimuli and timing of injury (Newman
and Reichenbach, 1996; Di Polo et al., 1998; Gauthier et al., 2005; Joly
et al., 2007; Pernet et al., 2007; Lebrun-Julien et al., 2009a,b, 2010;
Cueva Vargas et al., 2015, 2016). Most of our existing knowledge on
how glia regulate NVC in the visual system stems from studies focused
on the role of Müller glia (Newman, 2015; Biesecker et al., 2016; Nippert
and Newman, 2020), whereas not much is currently known about the
role of astrocytes in this response. Müller cells are ideally positioned to
act as intermediaries in NVC because their ne processes surround
retinal neurons and synapses as well as capillaries in all plexuses, which
can readily respond to vasoactive agents. Indeed, stimulation of Müller
cells in retinal explants evoked arteriole dilation demonstrating that
these glial cells release vasodilating factors (Metea and Newman, 2006a;
Mishra and Newman, 2010). Subsequent experiments showed that
light-evoked vasodilation is abolished when neuron-to-Müller glia
signaling is impaired. For example, when ATP receptors on Müller cells
were blocked using purinergic antagonists, light-evoked Ca
2+
increase
in Müller cells was eliminated thus preventing vessel dilation despite
persistent neuronal activity (Metea and Newman, 2006a). Light stimu-
lation increased Ca
2+
in Müller cell endfeet in contact with capillaries,
and capillary dilation was abolished when glial Ca
2+
signaling was
reduced (Biesecker et al., 2016). Although earlier studies reported a
correlation between increased Ca
2+
in Müller cells and vasodilation, the
current consensus is that Ca
2+
rises in Müller cells and brain astrocytes
exert dual effects - vasodilation or vasoconstriction - depending on
several parameters including pre-existing vessel tone, NO and O
2
levels
as well as the type of vasomodulators produced (Mulligan and MacVicar,
2004; Metea and Newman, 2006b; Attwell et al., 2010; Rungta et al.,
2016; Blanco et al., 2008; Chuquet et al., 2007).
Studies using isolated retina and brain slice preparations support a
model in which active neurons, with concomitant glutamate and/or ATP
release, signal Müller cells or astrocytes to increase Ca
2+
and produce
vasomodulators, which then act on vessels to regulate blood ow (Att-
well et al., 2010). Indeed, increased neuronal function boosts Ca
2+
levels
in Müller glia promoting the activation of phospholipase A
2
and syn-
thesis of arachidonic acid metabolites, which are potent vasomodulators
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
8
(Newman, 2015). The accumulation of arachidonic acid leads to the
production of metabolites such as prostaglandins (e.g. PGE
2
), epox-
yeicosatrienoic acids (EETs), and 20-hydroxy-eicosatetraenoic acid
(20-HETE), which can dilate or constrict nearby vessels (Zonta et al.,
2003; Metea and Newman, 2006b; Gordon et al., 2008). PGE
2
promotes
vasodilation by binding to the EP4 prostaglandin receptor (Yokoyama
et al., 2013), which stimulates adenylyl cyclase and increases cAMP
production to decrease intracellular Ca
2+
levels in mural cells (e.g.
smooth muscle cells and pericytes) (Mori et al., 2007; Misfeldt et al.,
2013). Conversely, PGE
2
can also induce vasoconstriction through
activation of the receptor for thromboxane, a vasoconstricting deriva-
tive of arachidonic acid, or the E prostanoid receptor-3 presumably by
decreasing cAMP levels (Abran et al., 1995; Wright and Harris, 2008;
Torring et al., 2014). EETs contribute to vasodilation by activating
Ca
2+
-dependent K
+
channels on vascular smooth muscle and endothelial
cells resulting in membrane hyperpolarization and relaxation (Campbell
and Fleming, 2010; Mori et al., 2021). Regarding the arachidonic acid
metabolite 20-HETE, this molecule was shown to promote light-evoked
vasoconstriction in isolated whole-mount retinas (Metea and Newman,
2006b). This nding is consistent with studies using hippocampal slices
showing that glial stimulation resulted in vasoconstriction mediated by
20-HETE (Mulligan and MacVicar, 2004).
In summary, glial cells in the retina respond rapidly to neuronal
activity by producing vasoactive agents including: i) prostaglandins,
which can exert either vasodilating or vasoconstricting responses, ii)
EETs which are vasodilating, and iii) 20-HETE which is vasoconstricting.
The limitations of these earlier studies is that they utilized whole-mount
preparations ex vivo, therefore little or no information from in vivo
studies exists. In addition, most of the studies examined the response of
retinal arterioles to glia-derived vasoactive factors. However, the
response of capillaries, which represent the vast majority of the retinal
microvasculature, remains poorly understood. Furthermore, there is
currently a lack of information about the role of Müller cell- or astrocyte-
derived vasomodulators in the context of glaucomatous damage,
through known or yet unknown factors, presenting a major gap in our
understanding of how glia contribute to NVC dysregulation in this
disease.
3.4.2. The role of microglia
Microglia are highly specialized resident macrophages ideally posi-
tioned to survey the central nervous system environment due to their
highly dynamic processes, plasticity, longevity, motility, and ability to
respond to many stimuli (Prinz et al., 2021). Microglia play critical roles
in the maintenance of tissue homeostasis during development, health,
and disease including neurogenesis, myelination, vasculogenesis, and
circuit renement (Hammond et al., 2018). The traditional view of
‘goodversus ‘bad microglia has been revised to include the multiple
variables that drive phenotypic transformation of microglia in myriad
conditions and at multiple levels including transcriptional, epigenetic,
translational, and metabolic (Paolicelli et al., 2022). Genetic depletion
of microglia expressing the C-X3-C motif chemokine receptor (CX3CR1)
did not alter the vascular pattern and laminar organization of the retina,
and retinal neurons did not undergo overt death (Wang et al., 2016).
However, the absence of microglia negatively affected synaptic trans-
mission and light-evoked retinal function (Wang et al., 2016). A recent
study showed that microglia depletion using PLX5622, a pharmacolog-
ical inhibitor of the colony-stimulating factor 1 receptor (CSF1R) that is
essential for microglia survival, exacerbated RGC loss during ocular
hypertension without compromising the pattern electroretinogram
(ERG) amplitude or visual acuity (Tan et al., 2022) supporting the
functional diversity of microglia. Consistently, studies on the role of
microglia in models of optic nerve injury including glaucoma have been
reported to be deleterious or benecial depending on the context. This
subject has been covered in recent reviews and will not be discussed here
(Ahmad and Subramani, 2022; Pitts and Margeta, 2023). Instead, we
will focus on the emerging role of microglia in the regulation of vascular
responses.
The role of microglia on NVC regulation and the mechanisms
involved are still poorly understood, but recent work suggests that these
cells interact with other members of the NVU to modulate blood ow.
Microglial processes are in dynamic contact with every cell type of the
NVU (Csaszar et al., 2022), making microglia ideal components to in-
uence and regulate vascular function. During development, microglia
play a critical role in vessel formation and modulate the sprouting,
migration, anastomosis and renement of the retinal vasculature
(Arnold and Betsholtz, 2013). Studies in which microglia were depleted
demonstrated that they play a key role in angiogenesis and the forma-
tion of new vessels (Checchin et al., 2006; Li et al., 2019) by secreting
soluble factors such as the Fas ligand (CD95L), which binds to its re-
ceptor on the endothelial cells lopodia (Chen et al., 2017). Further,
activation of the Notch signaling pathway by the MAS1 oncogene (MAS
receptor), the receptor of angiotensin-(17), has been shown to be
necessary for the recruitment of microglia around vessels to promotes
vascular growth in the developing retina (Foulquier et al., 2019; Outtz
et al., 2011). Microglia establish connections with pericytes and endo-
thelial cells early during development and, when pericytes are depleted,
the density and proliferation of microglia is markedly reduced (Hattori
et al., 2022) suggesting a critical crosstalk between microglia and
pericytes.
Microglia have been proposed to modulate vascular responses in a
context-dependent manner. For example, activation of the microglial
fractalkine-CX3CR1 signaling leads to capillary constriction through the
renin-angiotensin system, a pathway implicated in diabetic retinopathy
(Mills et al., 2021). Microglia depletion with a CSF1R inhibitor
(PLX3397) increased capillary diameter and cerebral blood ow (Bisht
et al., 2021). In contrast, a recent study reported that microglia ablation
impaired functional hyperemia by reducing cerebral blood ow
following whisker stimulation (Cs´
asz´
ar et al., 2022). In pathological
conditions, pathogen-activated microglia secrete pro-inammatory cy-
tokines that alter endothelial cell function (Rodríguez et al., 2022),
which is likely to disrupt neurovascular coupling. Some
microglia-vascular interactions are thought to be mediated by puriner-
gic signaling (Bisht et al., 2021; Cs´
asz´
ar et al., 2022), nonetheless, the
underlying mechanisms are far from being understood. The G-protein
coupled P2Y12 purinergic receptor (P2RY12) is expressed by microglia
(Zhang et al., 2014; Lou et al., 2016) and regulates microglial-directed
motility and migration in response to cell injury (Haynes et al., 2006).
Recent studies using conditional knockout mice for P2RY12 and the
purine channel pannexin 1 (PANX1), the main ATP release channel,
suggest that a microglia-dependent mechanism controls cerebral blood
ow (Bisht et al., 2021; Cs´
asz´
ar et al., 2022). Experiments in mice that
selectively targeted microglia by chemogenetics led to the disruption of
hemodynamic processes in response to ATP (Cs´
asz´
ar et al., 2022).
Moreover, in a model of cerebrovascular adaptation generated by
common carotid artery occlusion, microglia purinergic signaling was
necessary for normalization of cerebral blood ow, and microglia
depletion or blockade of P2RY12 led to decits in adaptation after
repeated vessel occlusion (Cs´
asz´
ar et al., 2022). Collectively, these
studies suggest that microglia may regulate NVC through purinergic
signaling. However, since purine and purinergic receptors are also
expressed by different cell types in the NVU, such as pericytes, endo-
thelial cells, and astrocytes (Hørlyck et al., 2021), further studies are
needed to unravel the precise mechanisms underlying the role of puri-
nergic signaling in blood ow regulation. Studies that directly assess the
role of microglia on NVC dysfunction in experimental glaucoma are
currently lacking.
4. Pericytes are key regulators of neurovascular coupling
4.1. How do pericytes regulate capillary blood ow and NVC?
Accumulating evidence using state-of-the-art approaches to
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
9
selectively manipulate pericytes in vivo demonstrate that these cells play
a critical role in the regulation of microcirculatory blood ow in both
retina and brain (Alarcon-Martinez et al., 2020, 2022; Hartmann et al.,
2022; Longden et al., 2023). Studies using optogenetics to selectively
stimulate brain pericytes showed that when these cells contract there is a
reduction in blood cell velocity and ux (Nelson et al., 2020; Hartmann
et al., 2021). In contrast, studies using optical ablation of cortical peri-
cytes led to the dilation of capillaries lacking pericyte coverage (Ber-
thiaume et al., 2018), a response that was also observed following
pericyte loss in a model of epileptic seizures (Arango-Lievano et al.,
2018). In the retina, laser-mediated ablation of specialized processes
connecting pericytes blocked NVC coupling (Alarcon-Martinez et al.,
2020), and pericyte contraction reduced capillary blood ow and NVC
regulation during glaucomatous damage (Alarcon-Martinez et al.,
2022). In a complementary study, optogenetic hyperpolarization of
retinal pericytes by selective expression of halorhodopsin, enhanced
capillary blood ow in vivo (Ivanova et al., 2021). Collectively, these
studies strongly support the view that pericytes play critical roles in the
hemodynamic regulation of capillary blood ow in physiological and
pathological conditions.
The nexus between neuronal activation and capillary responses has
been proposed to occur via receptors and channels located on pericytes.
Recent single cell-RNAseq analysis revealed that pericytes express
metabotropic, purinergic, prostanoid, chemokine, adenosine, and
adrenergic receptors; as well as K
+
, transient receptor potential, Ca
2+
,
and chloride channels (Vanlandewijck et al., 2018; Hariharan et al.,
2020; Yang et al., 2022). Neuronal- and glia-derived products can
activate these receptors/channels to initiate intracellular pathways
triggering membrane changes and Ca
2+
-dependent
α
-SMA variations to
promote pericyte contraction or relaxation, hence modulating capillary
diameter and blood ow (Puro, 2007; Peppiatt et al., 2006; Hamilton
et al., 2010; Hariharan et al., 2020). Based on this, the regulation of Ca
2+
levels within pericytes is essential to fulll their role as active regulators
of capillary hemodynamics. The primary pathways of Ca
2+
increase in
pericytes are voltage-dependent Ca
2+
channels (VDCC), the primary
gates of extracellular Ca
2+
into pericytes; and inositol 1,4,5-trisphos-
phate receptors (IP3Rs), which extrude Ca
2+
from the endoplasmic re-
ticulum to the cytosolic space (Burdyga and Borysova, 2018). VDCCs are
regulated by membrane potential changes and are typically composed of
distinct subunits (i.e.,
α
1
, β,
α
2
δ, and γ). Capillary pericytes are known to
express genes encoding the
α
subunits for L-type (Ca
v
1.2, Ca
v
1.3),
P/Q-type (Ca
v
2.1), and T-type (Ca
v
3.1, Ca
v
3.2) channels as well as genes
encoding the β and
α
2
δ auxiliary subunits (He et al., 2018; Vanlande-
wijck et al., 2018). In the retina, L-type VDCCs containing the Ca
v
1.2
subunit play critical roles in Ca
2+
signals that regulate
pericyte-mediated capillary constriction in experimental glaucoma
(Alarcon-Martinez et al., 2022). In addition, pericytes express transient
potential (TRP) channels (Vanlandewijck et al., 2018), a family of
non-selective ion channels located on the plasma membrane of many
cells that conduct signicant amounts of Ca
2+
(Per´
alvarez-Marín et al.,
2013; Ryskamp et al., 2011; Kriˇ
zaj et al., 2023). Among TRPs, the
vanilloid-type members TRPV1 and TRPV4 have been associated with
glaucomatous damage (Kriˇ
zaj et al., 2023). Indeed, genetic deletion of
TRPV1 reduced RGC somatic size, dendritic length as well as
complexity, and reduced spontaneous activity selectively in OFF
α
RGCs
(Risner et al., 2020) suggesting the involvement of TRPV1-mediated
pathways on RGC function. However, the role of TRP channels on per-
icyte function and their impact during glaucomatous damage is
currently unknown.
The predominant ion channel expressed by brain pericytes is the
ATP-sensitive K
+
(K
ATP
) channel, containing the inward rectier (K
ir
)
subunit K
ir
6.1, which accounts for nearly half of all ion channels
expressed by these cells (He et al., 2018; Vanlandewijck et al., 2018;
Hariharan et al., 2020; Sancho et al., 2022a; Ando et al., 2022). A recent
elegant study demonstrated that capillary pericytes act as metabolic
sentinels through K
ATP
channels, which respond to changing energy
levels, to regulate blood ow (Hariharan et al., 2022). Indeed, when
glucose is abundant and ATP levels are high in pericytes, K
ATP
channels
remain closed, but when glucose is low and ATP levels drop, K
ATP
channels open causing K
+
outow and pericyte membrane hyperpolar-
ization (Hariharan et al., 2022). The hyperpolarizing electrical signal
from pericytes is fed into the underlying endothelium, where it activates
endothelial cell K
IR
2.1 channels promoting upstream transmission to
dilate the penetrating arteriole and increase blood ow to replenish
glucose delivery (Hariharan et al., 2022).
The functional role of vasomodulators on the battery of receptors and
channels expressed by retinal pericytes remains poorly understood,
however, correlative information obtained by application of agonists
and antagonists has shed information on the response of pericytes. For
example, application of ATP or purinergic receptor agonists on isolated
whole retinas induced the contraction of pericytes soma around capil-
laries (Kawamura et al., 2002; Peppiatt et al., 2006) presumably through
a decrease in cAMP production via G protein signaling. Whether
ATP-induced pericyte contraction occurs via a direct or indirect mech-
anism is currently unknown. Arachidonic acid derivatives produced by
glial cells such as the prostaglandin PGE
2
can be further processed into
thromboxane A
2
, which can act on thromboxane receptors on pericytes
to promote contraction. In the retina, application of a synthetic analog of
PGE
2
led to marked pericyte contraction, which was blocked with in-
hibitors of actin depolymerization such as cytochalasin D and latrun-
culin B (Gonzales et al., 2020). Capillary pericytes also express high
levels of the ET-1 receptor A (ednra) (Vanlandewijck et al., 2018), and
display robust contraction in response to ET-1 (Zambach et al., 2021).
Although the mechanism by which ET-1 receptor signaling causes per-
icyte contraction has not been fully elucidated, it is presumed to be the
result of Ca
2+
increase in pericytes through opening of membrane
channels and/or intracellular stores leading to activation of their con-
tractile machinery. Studies in the retina suggest that ET-1 leads to per-
icyte contraction via multiple pathways including inhibition of
ATP-sensitive K
+
channels, which prevent hyperpolarization and peri-
cyte dilation, and reduced gap junction connectivity between pericytes
(Kawamura et al., 2002). In terms of pericyte relaxation leading to
vasodilation, several pathways have been identied including glutamate
(Hall et al., 2014) and adenosine, the latter likely promoting pericyte
relaxation by acting on A2a purine receptors (adora2) leading to
ATP-dependent K
+
channel ux (Li and Puro, 2001). These responses
require a high level of communication between pericytes and other cells
in the NVC unit as well as cross-talk between pericytes, which will be
described in the next section.
4.2. Discovery of inter-pericyte tunneling nanotubes (IP-TNTs)
The previous section covered a wealth of information supporting the
concept that pericytes regulate microcirculatory blood ow by con-
tracting and relaxing to induce changes in capillary diameter. However,
this model does not account for the need to coordinate dilation and
constriction across capillary networks to achieve the ne distribution of
blood supply required by active RGCs. This is particularly important in
the retina where patterns of visual stimulation imply that some neurons
are more active than others at any given time (Westheimer, 2007), thus
blood must be rapidly relocated to meet the demand of light-activated
versus light-inactivated neurons. Until recently, the mechanisms un-
derlying retinal blood distribution were poorly understood. Recent ad-
vances in this regard include the discovery of tunneling nanotube
(TNT)-like processes, which connect two bona de pericytes to form a
functional hemodynamic network in the mouse retina (Alarcon-Marti-
nez et al., 2020). These highly specialized microstructures, termed
inter-pericyte tunneling nanotubes (IP-TNTs), are extremely ne pro-
cesses that connect two distinct pericytes located on separate capillary
systems (Fig. 3). IP-TNTs emerge from the soma of a pericyte, named
proximal pericyte, and extend to a neighboring capillary where they
establish connections with a distal pericyte. They are abundantly
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
10
expressed in the mouse retina and optic nerve head as well as visual
centers in the brain (Fig. 4). Three-dimensional reconstruction revealed
that IP-TNTs are tubular, with an average diameter of ~500 nm that can
extend between 4
μ
m and 90
μ
m in length, and are abundant in all
retinal plexuses (Alarcon-Martinez et al., 2020). TNTs have been
described in other systems, including tumor and immune cells, and a
conserved structural requirement is that they contain F-actin (Yamashita
et al., 2018; Tarasiuk and Scuteri, 2022). Consistently, IP-TNTs are
endowed with an F-actin cytoskeleton as well as
α
-SMA, focal adhesion
kinase 1, and ribosomal protein S6, but they lack
α
-tubulin and
mesenchymal stem cell markers (Alarcon-Martinez et al., 2020).
IP-TNT-specic markers have yet to be identied.
TNTs have been proposed as a novel means for cell-cell communi-
cation and potential transfer of biological cargos between distant cells,
thus a phenotypic requirement is that they must connect two cells
(Mittal et al., 2019; Ortin-Martinez et al., 2021). Experiments using
single-pericyte electroporation demonstrated that low molecular weight
uorescent dyes (e.g. uorescein, 332 Da) diffused rapidly from the
proximal pericyte soma through the IP-TNT and into the soma of the
distal pericyte (Alarcon-Martinez et al., 2020) (Fig. 5A). In contrast,
larger molecular weight markers (e.g., dextran, 3 kDa) diffused within
the proximal pericyte, but did not enter the distal pericyte suggesting the
existence of gap junctions at this interface. When a low molecular
weight marker was electroporated into the proximal pericyte in the
presence of the gap junction blocker carbenoxolone (CBX), it accumu-
lated in the soma and IP-TNT (Fig. 5B). Dextran accumulation in the
proximal pericyte allowed unmistakable identication of two
IP-TNT-coupled pericytes as well as their connection at the level of the
IP-TNT terminal (end-foot) and the distal pericyte process
(Alarcon-Martinez et al., 2020). Ultrastructural analysis using focused
ion beam electron microscopy (FIB-SEM) further conrmed that IP-TNT
end-feet connect with distal pericyte processes through
membrane-to-membrane contacts evocative of gap junctions (Fig. 5C)
(Alarcon-Martinez et al., 2020). Consistent with this, expression of
connexin 43 (Cx43) was detected at IP-TNT end-feet (Alarcon-Martinez
et al., 2020) (Fig. 5D). Together, these data show that IP-TNTs are
close-ended processes in which the cytoplasm of the two linked pericytes
remain separate, but ions and small molecules can move through the gap
junctions. Of interest, other organelles were found within IP-TNTs
including endoplasmic reticulum, vesicles, and mitochondria, which
can move along the IP-TNT (Alarcon-Martinez et al., 2020), but are not
transferred to the distal pericyte because gap junctions at this interface
limit their passage.
How do IP-TNTs work? Using two-photon laser scanning microscopy,
Alarcon-Martinez et al. demonstrated that capillary pairs connected by
IP-TNTs always exhibit simultaneous, but opposite, responses triggered
by light stimulation: one capillary dilates while the other constricts
(Alarcon-Martinez et al., 2020). In other words, when the proximal
capillary dilates, the distal capillary constricts, and vice versa. Consis-
tent with caliber changes, IP-TNT-linked capillaries display opposite
blood ow responses: the dilating capillary increases blood ow, while
the constricting capillary reduces blood ow (Alarcon-Martinez et al.,
2020). Critically, targeted laser-induced ablation of individual IP-TNTs
completely impaired coordinated light-evoked changes in capillary
diameter and blood ow. Interestingly, matched responses between
IP-TNT-linked pericytes and capillaries was shown to be achieved
through intercellular Ca
2+
waves (ICWs). Live imaging of Ca
2+
tran-
sients in mice expressing the Ca
2+
sensor GCaMP6 selectively in peri-
cytes, demonstrated spontaneous synchronous Ca
2+
increases in
IP-TNT-coupled pericytes that propagated bidirectionally along IP-TNTs
(Alarcon-Martinez et al., 2020), similar to ICWs described in other TNTs
(Leybaert and Sanderson, 2012; Osswald et al., 2015). In addition,
light-evoked capillary dilation and constriction were coupled to oppo-
site and synchronized Ca
2+
transients in pericytes connected by
IP-TNTs. ICW frequency was substantially reduced after administration
of gap junction blockers (Alarcon-Martinez et al., 2020), which is
consistent with the observation that IP-TNTs connect with distal peri-
cytes via gap junctions, known to be required for ICW propagation
(Wang et al., 2010; Sherer, 2013). Furthermore, laser-mediated ablation
of IP-TNTs disrupted ICWs and abolished capillary responses as well as
blood ow regulation after light stimulation (Alarcon-Martinez et al.,
2020). Collectively, these ndings identify IP-TNTs as novel
nanotube-like processes that are essential for pericyte-to-pericyte
communication and NVC regulation in the living retina.
The discovery of IP-TNTs challenges previous simplistic models in
which light triggers only dilation to increase blood ow. Instead, the
current data support that IP-TNTs can actively participate in the com-
plex spatial and temporal heterogeneity of blood redistribution within
the retinal capillary network. It is plausible that IP-TNTs also regulate
blood ow and NVC within the ONH and RGC targets in the brain, given
their presence in higher visual centers (Alarcon-Martinez et al., 2020),
but this remains to be demonstrated. IP-TNTs are in close contact with
neurons and glia suggesting that IP-TNTs can be directly modulated by
vasoactive factors derived from these cells. Multiple lines of evidence
argue against IP-TNTs being merely empty sleeves from regressing
vessels or bridging pericytes (Franco et al., 2015; Korn and Augustin,
2015; Watson et al., 2017; Mendes-Jorge et al., 2012), and rather sup-
port their similarity with close-ended TNTs including: i) lack of endo-
thelial cell markers, ii) expression of
α
-SMA, iii) presence of organelles
including motile mitochondria, iv) process stability, and v) communi-
cation through bidirectional ICWs (Alarcon-Martinez et al., 2020). Many
questions remain regarding the functional role of IP-TNTs during
development, health, and disease. The next section discusses current
data on the role of IP-TNTs during ischemic and glaucomatous damage.
Fig. 3. Structure of inter-pericyte tunneling nanotubes (IP-TNTs). Schematic
representation of an IP-TNT, a highly specialized nanotube-like process that
connects two pericytes located on distant capillary systems. An individual IP-
TNT emerges from the soma of a parent pericyte, named proximal pericyte,
and extends to a neighboring capillary where it establishes a connection with
the process of a distal pericyte. IP-TNTs are tubular with an average diameter of
~500 nm, which can extend between 4
μ
m and 90
μ
m in length, and contain
organelles including abundant mitochondria. Created with BioRender.com.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
11
4.3. Pericytes and IP-TNTs during glaucomatous stress
In physiological conditions, pericytes and IP-TNTs play a critical in
vivo role in the regulation of local microvessel dynamics and NVC in the
adult retina (Alarcon-Martinez et al., 2020). However, what happens
when the system is under glaucomatous-related stress? Previous studies
demonstrated that pericytes can constrict capillaries in conditions of
stroke and ischemia, thus impairing capillary dynamics and preventing
blood ow during reperfusion (Yemisci et al., 2009; OFarrell et al.,
2017; Alarcon-Martinez et al., 2019). More recently, live imaging
studies showed that pericytes constrict capillaries during ocular hyper-
tension, decreasing blood supply and compromising RGC function
(Alarcon-Martinez et al., 2022). Indeed, a substantial reduction in
capillary diameter and blood ow at pericyte locations was detected in a
mouse model of microbead-induced IOP elevation in mice, prior to overt
RGC death (Alarcon-Martinez et al., 2022). Furthermore, two-photon
microscope live imaging of capillary dynamics and blood ow, recor-
ded before and after light stimulation, showed that the ability of retinal
capillaries to dilate in response to light was severely compromised
during glaucomatous damage (Alarcon-Martinez et al., 2022). Of in-
terest, the probability of capillary blood ow blockage at pericyte lo-
cations also increased several-fold in eyes subjected to ocular
hypertension relative to sham-operated controls (Alarcon-Martinez
et al., 2022).
Consistent with their role in NVC, pathological stress has been shown
to provoke structural and functional alterations in IP-TNTs. Recent work
reported that a substantial number of IP-TNTs, both in the retina and
optic nerve, were ruptured in conditions of elevated IOP (Alarcon--
Martinez et al., 2022). A similar loss of IP-TNT integrity was observed
during ischemia or glucose deprivation (Alarcon-Martinez et al., 2020).
In these studies, IP-TNT breakage was accompanied by a substantial loss
of light-evoked capillary responses, notably impaired dilation, the
inability to regulate blood ow, and a substantial decrease in the fre-
quency of ICWs. There is now increasing evidence that Ca
2+
homeostasis
in pericytes plays a critical role in the maintenance of IP-TNT structure
and function. In addition to playing a key role in pericyte-to-pericyte
communication through ICWs, cytosolic Ca
2+
regulates the contractile
activity of pericytes (Burdyga and Borysova, 2018). Using transgenic
mice that express the Ca
2+
sensor GCaMP6f selectively in pericytes, a
recent study demonstrated that ocular hypertension induced a robust
and sustained increase of Ca
2+
within pericytes and their IP-TNTs
(Alarcon-Martinez et al., 2022). Ca
2+
increase in pericytes was also
observed after transient ischemia and correlated with reduced frequency
of ICWs in IP-TNT-coupled pericytes (Alarcon-Martinez et al., 2020). A
major pathway of extracellular Ca
2+
entry into pericytes is through
L-type VDCCs (Borysova et al., 2013), with the alpha 1C subunit
Fig. 4. IP-TNTs form a network of connected capillaries in all vascular plexuses of the retina and in the optic nerve. (A) Visualization of IP-TNTs in retinas from mice
expressing red uorescent protein under the control of the NG2 (Cspg4) promoter (NG2-DsRed) for selective imaging of retinal pericytes. Laminin (green) is used to
label capillaries. In healthy retinas, IP-TNTs (white arrowheads) form networks linking pericytes on separate capillary systems. Endogenous DsRed within the IP-TNT,
conrming its pericyte origin, can be seen at higher magnication (inset A
). (B) Analysis of the optic nerve head (ONH) in an NG2-DsRed mouse shows a network of
IP-TNTs connecting pericytes between capillaries (white arrowheads). Endogenous DsRed uorescence in the IP-TNT is shown in the higher magnication inset (B
).
(C) IP-TNTs are detected in the visual cortex and can be visualized in xed tissue or by live imaging using two-photon laser scanning microscopy (TPLSM). Modied
from (Alarcon-Martinez et al., 2020, 2021).
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
12
(Cav1.2) of these Ca
2+
channels particularly enriched in pericytes
(Vanlandewijck et al., 2018). Importantly, pericyte-specic conditional
deletion of the gene encoding Cav1.2 (Cacna1c) had several benecial
effects during ocular hypertension including: i) protection of IP-TNTs, ii)
restoration of capillary dynamics, blood blow regulation, and NVC, iii)
recovery of RGC function, and iv) enhancement of RGC survival (Alar-
con-Martinez et al., 2022). Collectively, these ndings indicate that
maintaining Ca
2+
homeostasis in pericytes is critical to restore vascular
and neuronal function in glaucoma and prevent RGC death. In addition,
these studies identify pericytes and IP-TNTs as central components of the
NVU with therapeutic potential to prevent or reverse vascular dysre-
gulation in glaucoma.
4.4. RGC-pericyte crosstalk: the chicken or the egg?
The traditional view of neurovascular interactions in glaucoma is
that, as RGCs die, there is less demand for oxygen and energy leading to
reduced local blood ow in areas of neurodegeneration. This model is
supported by studies in retina and brain showing that neurons initiate
local neurovascular response by signaling to blood vessels, directly or
indirectly, as discussed in previous sections. In this scenario, unbalanced
delivery of neuronal-derived products upon pericyte receptors may lead
to abnormal vascular function and blood delivery that will, in turn, seal
the fate of an injured neuron. Recent evidence, however, suggests that
reverse signaling from pericytes/capillaries to RGCs may also contribute
to NVC pathology in glaucoma. In this regard, a recent study used
multiphoton microscopy to longitudinally capture single-RGC Ca
2+
re-
sponses along with blood ow in the capillary serving the same neuron
before and after pericyte-induced vessel constriction (Alarcon-Martinez
et al., 2022). This work demonstrated that when retinal capillary blood
ow was within the normal range, light-evoked RGC Ca
2+
responses
were robust and decayed rapidly; but when blood ow was compro-
mised, Ca
2+
signals were reduced and recovery was signicantly
delayed (Alarcon-Martinez et al., 2022). These ndings suggest that
pericyte/capillary dysfunction directly impairs neuronal activity and
may compromise RGC viability in glaucoma. In line with this reversal of
roles, the hemodynamic response has been shown to inuence neuronal
activity through mechanical, thermal, or chemical effects on astrocytes,
also known as the hemo-neural hypothesis or vasculo-neuronal coupling
(Kim et al., 2016; Moore and Cao, 2008). For example, when the
transmural pressure/ow in penetrating arterioles was increased in
brain slices, a concomitant suppression of pyramidal cell activity was
observed through activation of mechanosensitive TRPV4 channels in
brain astrocytes (Kim et al., 2016). Collectively, these data suggest an
autoregulatory mechanism in which intravascular pressure, and poten-
tially eye pressure through vascular responses, could directly modulate
neuronal activity. The implications and mechanisms underlying these
observations require further investigation.
5. Alterations of blood-retinal/brain barriers in glaucoma
In addition to being essential for the regulation of NVC in the retina
and optic nerve, the cells of the NVU play critical roles in maintaining
the integrity of the blood-retina- and blood-brain-barriers (BRB and
BBB, respectively) (OLeary and Campbell, 2023; Moyaert et al., 2023).
The microenvironment in the retina and optic nerve is tightly regulated
and kept separate from the systemic circulation by the BRB and the BBB,
which prevent the entry of cytotoxic plasma components, blood cells,
and pathogens (Montagne et al., 2017). At the same time, these barriers
nely regulate the transport of molecules to maintain a tightly
Fig. 5. IP-TNTs connect two bona de pericytes via CX43 gap junctions. (A) Electroporation of a single pericyte with the low molecular weight marker uorescein
shows rapid diffusion of the dye from the soma and IP-TNT into the distal pericyte demonstrating a direct connection between IP-TNT-linked pericytes (pipette shown
in green). (B) When uorescein is electroporated in the presence of the gap junction blocker carbenoxolone (CBX), the dye accumulates in the proximal pericyte and
IP-TNT delineating the contact between the IP-TNT end-foot (ef) shown in green, and the distal pericyte process (dpp) shown in red (see insets). Fluorescein
accumulation in the proximal pericyte allows unmistakable identication of the connection between the IP-TNT terminal and the distal pericyte. (C) Correlative serial
block-face focused ion beam scanning electron microscopy (FIB-SEM) revealed that IP-TNT end-feet (ef) connect with the distal pericyte process (dpp) through direct
membrane-to-membrane contacts, evocative of gap junctions (white arrowheads). (D) Consistent with this, connexin 43 (Cx43) is detected in IP-TNT terminals (end-
foot: ef) in Cx43-ECFP reporter mice (white arrowheads). Lectin is used to label pericytes, IP-TNTs, and endothelial cells. Modied from (Alarcon-Martinez
et al., 2020).
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
13
controlled milieu that is required for optimal tissue homeostasis and
neuronal activity (Zhao et al., 2015b). The outer BRB consists of the
choroid, Bruchs membrane, and retinal pigment epithelium, while the
inner BRB is demarcated by the endothelial cells that line the retinal
vasculature originating from the retinal artery. The properties of the
inner BRB are dened by the cells of the NVU, of which pericytes,
endothelial, and glial cells play key roles (Sweeney et al., 2019), and will
be discussed in subsequent sections. For the purpose of this review and
in the context of glaucoma, the term BRB will be used solely for the inner
BRB. The vascular barrier at the ONH and optic nerve is an intrinsic part
of the central nervous system and thus considered very similar in
structure and function to the BBB (Nian et al., 2021). In disease states,
including glaucoma, BRB/BBB breakdown can result in the leakage of
harmful blood components into the retina and optic nerve leading to
inammation, cellular inltration, aberrant transport and clearance of
molecules, reduced blood ow, and neuronal decits (Zhao et al.,
2015b; OLeary and Campbell, 2023). In the following sections, we will
cover the clinical evidence supporting alterations of the BRB/BBB
function in glaucoma patients as well as cellular and molecular mech-
anisms involved in the regulation of vascular permeability.
5.1. Loss of vascular integrity in glaucoma
5.1.1. Fluorescein angiography studies
Accumulating evidence from pre-clinical and clinical studies using
uorescein, a recognized physiological marker of the integrity of the
BRB/BBB function (Cunha-Vaz, 2004), suggest vascular barrier disrup-
tion in glaucoma. Experimental IOP elevation in non-human primate
eyes demonstrated uorescein extravasation at the optic disc (Radius
and Anderson, 1980). A number of clinical studies also reported uo-
rescein lling defects and increased leakage at the optic disc in glau-
coma relative to normotensive individuals (Arend et al., 2004, 2005;
Ashworth and Rosen, 1970; Plange et al., 2006, 2010, 2012; Schwartz,
1994; Schwartz et al., 1977; Spaeth, 1975; Tsukahara, 1978). Fluores-
cein dye extravasation is a dynamic process and, as such, the relative
increase of the late phase of uorescein leakage may suggest higher
vascular permeability at the optic nerve, which could stem from damage
to the BBB (Anderson and Davis, 1996; Orgül et al., 1999). For example,
the evaluation of the time course of uorescein leakage of the ONH in
glaucoma patients revealed an increase from 78 min to 910 min,
which was signicantly higher relative to healthy controls (Plange et al.,
2010). In addition, Arend et al. identied greater leakage in glaucom-
atous eyes using a semiquantitative digital image analysis in late-phase
angiogram (Arend et al., 2005). Leakage worsened in areas of peri-
papillary choroidal atrophy, a well-known feature of glaucomatous optic
neuropathy (Schwartz, 1994; Schwartz et al., 1977; Spaeth, 1975; Tsu-
kahara, 1978). Using uorescein angiography with a scanning laser
ophthalmoscope and semi-quantied digital image analysis, Plange
et al. identied increased uorescein leakage at the optic disc in patients
with high IOP (Plange et al., 2010). However, no association was found
with the extent of glaucomatous damage, either in visual eld damage or
cup-disc ratios (Plange et al., 2010), suggesting that glaucomatous
uorescein leakage may occur spatially and temporally regardless of
disease severity.
Fluorescein angiography has also revealed prolonged arteriovenous
transit times, with frequent vessel lling defects or delayed lling in
glaucomatous eyes with disc hemorrhages, suggesting vascular stasis
and thrombus formation at disc hemorrhage sites as a consequence of
vascular leakage (Park et al., 2015). Absolute and relative uorescein
lling defects of the ONH have been reported in glaucomatous eyes
(Schwartz et al., 1977; Spaeth, 1975; Talusan and Schwartz, 1977).
Absolute lling defects are larger and more frequent in patients with
glaucoma compared with healthy controls and correlate with capillary
dropout in the surface nerve ber layer of the optic disc (Loebl and
Schwartz, 1977; Nanba and Schwartz, 1988; Schwartz et al., 1977).
Fluorescein lling defects are preferentially located at the margin of the
optic disc excavation, predominantly at inferotemporal and super-
otemporal locations, and are often found at the wall rather than at the
oor of the cup (Adam and Schwartz, 1980; Fishbein and Schwartz,
1977; Schwartz, 1994; Schwartz et al., 1977; Arnold, 1995). The relative
defects are characterized by delayed uorescence, and they are inter-
preted as areas of hypovascularity. A correlation between the number,
extent, and topography of uorescein lling defects with visual eld
loss, retinal nerve ber layer defects, and excavation of the disc has been
reported in glaucoma patients (Nanba and Schwartz, 1988; Talusan and
Schwartz, 1977; Tsukahara, 1978).
5.1.2. Disc hemorrhages
An association between glaucoma and optic disc hemorrhages was
rst described in 1876 by Albert Emmerich, who reported bleeding
within the ONH and retina in some glaucoma patients (Emmerich,
1876). It was not until 1970 that Stephen Drance and Ian Begg found an
association between disc hemorrhages and visual eld defects, sug-
gesting that they are important pathological features in individuals
affected by glaucoma (Drance and Begg, 1970; Begg et al., 1970). Disc
hemorrhages are small splinter-like or ame-shaped areas of bleeding
located in the neuroretinal rim of the optic disc and the adjacent su-
percial nerve ber layer (Drance, 1989). Accumulating clinical evi-
dence conrms that optic disc hemorrhages are a signicant risk factor
for the development and progression of glaucoma and can be predictive
of future damage in patients with both primary open-angle and
low-tension glaucoma (Budenz et al., 2017; Jasty et al., 2020).
The disruption of the integrity of the capillary wall at the ONH is
considered to be at the origin of disc hemorrhages. Although the
mechanisms underlying the formation of disc hemorrhages are still
unknown, three main hypotheses have been proposed including
vascular defects, biomechanical stress, and reactive gliosis (Jasty et al.,
2020). Vascular dysregulation and alterations in NVU components can
lead to abnormal vessel constriction, microinfarction in the optic disc,
and decreased capillary perfusion (Drance and Begg, 1970; Grieshaber
et al., 2007; Lichter and Henderson, 1978), which increase the vulner-
ability of capillaries to rupture. In addition, the release of vasoactive
agents, cytokines, and mediators that increase BRB/BBB permeability
such as ET-1 and metalloproteinase-9 (MMP-9) may weaken the base-
ment membrane surrounding the capillary walls resulting in leaky ves-
sels (Mi et al., 2012; Lee et al., 2014). Microvascular ischemia and
autoregulatory dysfunction have also been suggested to play a role in the
development of disc hemorrhages (Grieshaber et al., 2006). Biome-
chanical strain, including sheer and compression forces, are known to
cause deformation of the lamina cribrosa as well as the prelaminar rim
tissue and can lead to microvascular disruption, leaky capillaries,
disturbance of the BRB/BBB, and hemodynamic defects (Quigley et al.,
1981, Burgoyne et al., 2005; Kim et al., 2015; Lee et al., 2022). Posterior
stress/strain forces at the lamina cribrosa may also affect vascular
endothelial cell differentiation, maturation, and molecular signaling
which may alter endothelial cell morphology, gene expression, and
function (Akimoto et al., 2000; Ott et al., 1995). Lastly, proliferative
reactive gliosis has been proposed to promote glial scar formation, hence
remodeling the ONH environment, causing breakdown of capillary walls
that result in optic disc hemorrhages (Lee et al., 2017a). Consistent with
this, reactive astrocytes produce several mediators known to open the
BRB/BBB including ATP, glutamate, aspartate, taurine, and
macrophage-inammatory protein 2 (Abbott, 2002; Chen et al., 2000;
Kustova et al., 1999). A better understanding of the origin of disc
hemorrhages may provide insights into the role of vascular leakage in
glaucoma.
5.2. Pericytes and the regulation of vascular permeability
In addition to the critical role of pericytes in the control of capillary
blood ow and NVC discussed in previous sections, these cells are
crucial for the maintenance of vascular barriers and their integrity
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
14
(Armulik et al., 2010; Daneman et al., 2010; Bell et al., 2010). Pericytes
are required to develop and maintain the homeostasis of the BRB/BBB
(Trost et al., 2016; OLeary and Campbell, 2023). For example, pericytes
are a key source of basement membrane proteins at the BRB/BBB such as
laminins and vitronectin (Yao et al., 2014; He et al., 2016) and they are
important players in endothelial tube formation and stabilization
(Armulik et al., 2011). During vascular development, pericytes are
recruited by platelet-derived growth factor B (PDGF-B), produced by
endothelial cells, which activates the PDGF receptor-β (PDGFR-β) on
pericytes (Park et al., 2017). Blockade of PDGF-B/PDGFRβ signaling in
mice using multiple approaches including conditional deletion of the
PDGFB gene (Enge, 2002; Park et al., 2017) or its retention motif
(Lindblom et al., 2003) and administration of PDGFRβ blocking anti-
bodies (Uemura et al., 2002; Ogura et al., 2017) resulted in vascular
barrier disruption characterized by reduced pericyte coverage,
increased BRB/BBB permeability, and microaneurysms (Armulik et al.,
2010; Daneman et al., 2010; Bell et al., 2010; Lindahl et al., 1997).
Analysis of mice with different combinations of null, hypomorphic, and
wild-type PDGFRβ alleles showed that the permeability of the BBB
correlated inversely with the amount of pericyte vessel coverage,
demonstrating that the number of pericytes directly determines vessel
permeability during development (Daneman et al., 2010).
In the retina, inhibition of pericyte recruitment using a PDGFRβ
blocking antibody promoted the dissociation of pericytes from endo-
thelial cells and BRB breakdown in adult mice reproducing classical
features of diabetic retinopathy such as increased vessel leakage,
hypoperfusion, and angiogenesis (Ogura et al., 2017). In this study,
transcriptomic analysis of pericyte-depleted endothelial cells showed
upregulation of inammatory genes, which correlated with an inux of
inltrating leukocytes intimately associated with the denuded vessels
(Ogura et al., 2017). Tamoxifen-induced conditional deletion of PDGFB
from developing endothelial cells conrmed that impaired pericyte
recruitment to retinal vessels leads to BRB disruption and diabetic
retinopathy-like features (Park et al., 2017). Despite retinal pericyte
loss, however, tamoxifen treatment in adult mice did not cause BRB
disintegration unless endothelial cells were challenged with exogenous
vascular endothelial growth factor-A (VEGF-A), which led to upregula-
tion of angiopoietin-2 (Ang2) and BRB breakdown (Park et al., 2017).
Thus, in addition to PDGF-B/PDGFRβ signaling, the interaction between
pericytes and endothelial cells involves other pathways including
Ang2/Tie-2 receptor signaling, transforming growth factor-β (TGF-β),
and Notch receptors and their ligands (Sweeney and Foldes, 2018).
These ndings support a tight pericyte-endothelial cell crosstalk in the
regulation of BRB/BBB integrity that involves paracrine mediators as
well as changes in gene expression (Geevarghese and Herman, 2014;
M¨
ae et al., 2021).
BRB/BBB breakdown secondary to pericyte depletion eventually
leads to neuronal dysfunction and functional cognitive impairment in
prevalent pathologies such as diabetic retinopathy and Alzheimers
disease (Yamazaki and Kanekiyo, 2017; Uemura et al., 2020; OLeary
and Campbell, 2023; Nikolakopoulou et al., 2019). However, there is no
evidence of pericyte death in glaucoma suggesting that other mecha-
nisms of pericyte dysfunction can potentially contribute to BRB/BBB loss
of integrity in this disease. A number of mediators could participate in
pericyte-induced BRB/BBB breakdown. For example, using a model of
ischemic damage induced by photothrombotic occlusion of cortical
capillaries, Underly and colleagues showed that capillary leakage ap-
pears at sites where pericyte soma are in contact with endothelial cells
(Underly et al., 2017). Plasma leakage during cerebral ischemia corre-
lated with MMP activation in pericytes, detected using a FITC-gelatin
probe MMP-2/9 activity (Underly et al., 2017). Injection of a MMP-9
inhibitor reduced pericyte-associated FITC-gelatin uorescence and
plasma leakage in the BBB (Underly et al., 2017). In the retina, MMP-9
activity has long been linked to RGC death in several models of retinal
damage, including ocular hypertension (Guo et al., 2005), optic nerve
transection (Sun et al., 2011), optic nerve ligation (Zhang et al., 2004b;
Zhang and Chintala, 2004), and kainic acid excitotoxicity (Zhang et al.,
2004a). Whether pericytes contribute to glaucoma pathophysiology by
BRB/BBB dysfunction caused by MMP proteolytic degradation or by
other mechanisms remains unknown.
5.3. Tight and adherens junctions in endothelial cells
Unlike the leaky capillary endothelium in peripheral organs, endo-
thelial cells that line the BRB/BBB are sealed by tight junctions and have
low rates of transcytosis (Reese and Karnovsky, 1967; Tietz and Engel-
hardt, 2015; Siegenthaler et al., 2013; Daneman and Prat, 2015).
Indeed, endothelial cells with tight and adherens junctions are a major
barrier keeping the retinal microenvironment closed off from the gen-
eral circulation (Nitta et al., 2003; Fresta et al., 2020). Tight junctions,
located near the endothelial cell apical membrane, include claudins (1,
3, 5, and 12), occludin, and junctional adhesion molecules, which limit
the paracellular diffusion of ions and solutes (Tietz and Engelhardt,
2015). Claudin-5 is a critical determinant of BBB/BRB permeability, and
loss of claudin-5 function has been shown to increase vascular leakage in
both retina and brain (Nitta et al., 2003; Argaw et al., 2009; Tian et al.,
2014; Muthusamy et al., 2014; Greene et al., 2019). Downregulation of
claudin-5 was reported in retinas from mice subjected to hypoxia, which
correlated with increased BRB permeability, similar to the phenotype
seen in claudin-5 decient mice (Koto et al., 2007). Claudin-5 down-
regulation was reported in a mouse model of acute ocular hypertension
(Zhao et al., 2020). Claudins can also be used therapeutically to seal the
BBB as shown in a model of experimental autoimmune encephalomy-
elitis by increasing the expression of claudin-1 (Pfeiffer et al., 2011).
Tight junction proteins are linked to the actin and vinculin-based cyto-
skeleton through scaffolding zona occludens proteins of the
membrane-associated guanylate kinase family (ZO-1, -2, and -3) (Tor-
navaca et al., 2015). ZO-1 deciency can contribute to BBB breakdown
in chronic and acute neurodegenerative disorders (Zlokovic, 2011). Of
interest, pericyte deciency or dysfunction can cause a reduction in the
expression of tight junction proteins by endothelial cells including
claudin-5, occludin, and ZO-1 (Bell et al., 2010), which increase ow
transcytosis across vascular barriers (Armulik et al., 2010).
The adherens junction proteins, vascular endothelial-cadherin (VE-
cadherin) and platelet endothelial cell adhesion molecule-1 (PECAM-1),
form homophilic cell-to-cell contacts near the basolateral membrane of
endothelial cells (Vorbrodt and Dobrogowska, 2003; Dejana and Vest-
weber, 2013; Tietz and Engelhardt, 2015). Adherens junctions connect
to the cytoskeleton, modulate receptor signaling, and regulate the
trans-endothelial passage of immune cells including monocytes, neu-
trophils, and lymphocytes (Giri et al., 2000; Allport et al., 2000; Tur-
owski et al., 2008; Wessel et al., 2014). In the retina, loss of VE-cadherin
was observed in mouse retinal organotypic cultures exposed to elevated
eye pressure (Brockhaus et al., 2020). However, the phosphorylation of
VE-cadherin at tyrosine residues was shown to be required for the
regulation of trans-endothelial migration of leukocytes in the brain
(Turowski et al., 2008; Wessel et al., 2014). Therefore, in addition to
changes in gene and protein expression, the analysis of VE-cadherin
phosphorylation would provide useful information about its contribu-
tion to vascular leakage in glaucoma. Overall, the functional role of tight
and adherens junctions in the onset and progression of BRB/BBB
breakdown in glaucoma remains to be elucidated.
5.4. Connexins and pannexins
Connexins and pannexins are transmembrane proteins that allow the
exchange of ions and molecules allowing autocrine and paracrine
signaling between cells. Connexin proteins form both connexin gap
junction channels and hemichannels. While gap junctions are required
for a number of physiological and neuroprotective functions, increasing
evidence suggests that hemichannels exert a pathological and pro-
degenerative role during injury or disease (Danesh-Meyer et al., 2016;
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
15
Boal et al., 2021). Connexin gap junction channels mediate cell-to-cell
intercellular communication and signaling (Willebrords et al., 2016)
by facilitating the passive diffusion of ions and metabolites between
adjacent cells to regulate electrical and chemical transmission (Weid-
mann, 1969). Connexin hemichannels, on the other hand, are cyto-
plasmic non-selective membrane pores that allow the passage of
molecules up to ~1 kDa in size (Yang et al., 2023). Due to their poor
selectivity and massive opening, which allows the passage of toxic me-
tabolites, the function of connexin hemichannels has been associated
with pathological conditions (Guo et al., 2016). Connexin hemichannels
are considered pathological pores primarily due to their low opening
probability in physiological conditions and large opening probability in
response to cellular stress (Contreras et al., 2003; Lander et al., 1996;
Giaume et al., 2013; Acosta et al., 2021). Consequently, dysregulated
connexin hemichannel opening has been shown to promote cell death
because of loss of osmoregulation, excitotoxicity, and inammation
(Yang et al., 2023).
Pannexins are also cytoplasmic channels permeable to small mole-
cules including Ca
2+
, glutamate, and ATP, but they differ from con-
nexins in that they rarely form intercellular channels (Cotrina et al.,
1998; Wei et al., 2016b). Although pannexins and connexins are struc-
turally similar, they do not share enough sequence similarity to have a
common ancestry (Beyer and Berthoud, 2018). The mechanisms that
trigger connexin hemichannel opening can also activate pannexin
channels including mechanical stimulation and Ca
2+
increase (Wei
et al., 2016b; Cotrina et al., 1998; Lohman and Isakson, 2014). Pannexin
channels are self-regulated and they close in response to extracellular
ATP, an important trigger during retinal injury (Barbe et al., 2006).
Recent evidence highlights the time-dependent roles of connexin hem-
ichannel and pannexin channels during ischemia (Mugisho et al., 2018).
It has been proposed that pannexin channels initiate the early response
to ischemic injury, and connexin hemichannels perpetuate the response
in sustained pathological conditions (Bernstein and Fishman, 2016;
Davidson et al., 2013).
Connexin 43 (CX43) is the most ubiquitous of all connexins and plays
an important role in several retinal injuries and diseases (Danesh-Meyer
et al., 2016; Boal et al., 2021). CX43 hemichannels are present in
endothelial cells, pericytes, Müller cells/astrocytes in both retina and
optic nerve of mice and humans (Janssen-Bienhold et al., 1998; Kerr
et al., 2010), thus they are key elements for crosstalk within the NVU.
IP-TNTs in the retina connect distal pericytes through their specialized
endfeet processes rich in CX43, which is essential for Ca
2+
-based
communication (Alarcon-Martinez et al., 2020). CX43 expression has
been shown to increase in an organotypic model of optic nerve ischemia,
and antisense oligonucleotide treatment substantially reduced CX43
levels and the ensuing inammatory response (Danesh-Meyer et al.,
2008). Partial optic nerve transection evoked a biphasic CX43: rapid
upregulation in the retinal area corresponding to axonal damage and
delayed elevation in the unaffected area, which coincided with signi-
cant RGC loss (Chew et al., 2011). A study of human post-mortem eyes
demonstrated a signicant increase in CX43 labeling in the lamina cri-
brosa of glaucoma patients (Kerr et al., 2010, 2011). In response to
cellular stress, CX43 hemichannels mediate ATP release into the extra-
cellular space (Cotrina et al., 1998; Iglesias et al., 2009; Liu et al., 2008),
which activates the purinergic inammatory pathway by binding to the
purinergic receptor, P2RX7 (Calder et al., 2015; Cotrina and Neder-
gaard, 2009; Huang et al., 2012). Extracellular ATP release via CX43
hemichannels was conrmed using CX43 knockout mice in which this
response was abrogated (Huang et al., 2012).
Accumulating evidence support a role for CX43 hemichannels in
vascular leakage in the retina and optic nerve. Mechanical stress and
strain has been shown to increase CX43 hemichannel expression leading
to ATP release from ONH astrocytes (Bao et al., 2004; Beckel et al., 2014;
Dubyak, 2009). In a retinal ischemia-reperfusion injury model, treat-
ment with the non-specic gap junction channel blocker carbenoxolone,
the hemichannel blocker lanthanum chloride, or CX43 hemichannel
mimetic blocking peptides prevented endothelial cell death, restored
vascular integrity, and promoted RGC survival (Danesh-Meyer et al.,
2012). These results were conrmed ex vivo in preparations of rat and
human microvascular endothelial cells exposed to hypoxia and treated
with CX43 blocking peptides (Danesh-Meyer et al., 2012) used at a
concentration that blocks hemichannels but does not uncouple gap
junctions (OCarroll et al., 2008). In this study, endothelial cell
dysfunction was mediated by opening of CX43 hemichannels in endo-
thelial cells in the absence of astrocytes (Danesh-Meyer et al., 2012).
This nding suggests that ischemia can cause CX43
hemichannel-mediated loss of endothelial cell integrity without the
involvement of astrocytes. High pressure-induced retinal
ischemia-reperfusion also resulted in upregulation of CX43 expression in
endothelial cells and astrocytes in the retinal nerve ber layer, which
correlated with BRB breakdown (Kerr et al., 2012). The role of CX43 in
BRB breakdown was conrmed using a CX43 hemichannel blocking
peptide, which decreased BRB disruption and neuroinammation (Chen
et al., 2015; OCarroll et al., 2013). Collectively, these ndings suggest
that the upregulation of CX43 hemichannels induced by ischemic injury
is an initiating event responsible for vascular leakage and an inam-
matory cascade leading to RGC death. The modulation of CX43 hemi-
channel opening has also been shown to effectively restore vascular
integrity in models of perinatal ischemia, spinal cord injury, and
light-induced macular damage (OCarroll et al., 2008; Guo et al., 2016;
Mao et al., 2017; Galinsky et al., 2018; Mat Nor et al., 2020). These
studies open up the possibility of using strategies to modulate CX43
function to prevent BRB/BBB breakdown and protect RGCs from
ensuing damage.
6. Conclusions and future directions
Accumulating evidence supports a connection between neuro-
vascular dysfunction and visual decits in glaucoma patients. The na-
ture of vascular defects is complex and includes impaired
autoregulation, NVC dysfunction, and loss of BRB/BBB integrity. An
imbalance in the regulation of microcirculatory blood ow and vascular
breakdown can increase the susceptibility of vulnerable RGCs to
stressors leading to neurodegeneration. Cell-cell interactions within
members of the NVU is critical for hemodynamic regulation and barrier
maintenance. Recent data suggest that signaling between NVU cells is
altered in glaucoma, but our current understanding of these processes
and how they relate to RGC dysfunction is still in its infancy. Due to their
strategic location, pericytes are emerging as key players in the regula-
tion of blood ow and NVC in the retina and optic nerve both in phys-
iological and glaucomatous conditions. Pericytes communicate with
each other via IP-TNTs, which provide a physical nexus for the coordi-
nation of light-evoked blood ow within a local capillary network. IP-
TNT damage triggered by ocular hypertension results in loss of NVC
and capillary responses, while strategies that protect IP-TNTs restore
blood ow promoting RGC survival and functional recovery. A better
understanding of how pericytes interact with other cells of the NVU will
provide insights into the causes of NVC dysfunction and will expand the
identication of potential therapeutic targets to restore hemodynamic
homeostasis in glaucoma. Unlike neurons, which have a limited capacity
for regeneration, vascular cells are plastic and contain self-repair
mechanisms thus presenting a unique opportunity to develop strate-
gies to support neurovascular function and maintain RGC health.
Author statement
LAM: conceptualization, data curation, formal analysis, investiga-
tion, methodology, writing - original draft, visualization; YS: data
curation, formal analysis, investigation, methodology, writing - original
draft, visualization; DVB: data curation, formal analysis, investigation,
methodology, writing - original draft, visualization; JLCV: writing -
original draft; IAVP: writing - original draft; HQ: writing - original draft,
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
16
visualization; BF: writing - original draft, HDM: writing - original draft;
ADP: conceptualization, data curation, formal analysis, writing orig-
inal draft, funding acquisition, supervision. All authors: reviewing and
editing of the manuscript.
Permission was obtained for the use of copyrighted gures from
other sources. The reference is included in the gure legend.
Declaration of competing interest
The authors declare that they have no competing nancial interest
regarding the content of this work.
Data availability
Data will be made available on request.
Acknowledgements
We thank Dr. Lin Wang for valuable comments on the manuscript.
Funding: Canadian Institutes of Health Research (ADP), National In-
stitutes of Health (ADP, BF), Glaucoma Foundation (ADP), BrightFocus
Foundation (ADP), Alcon Research Institute (ADP, LAM), and Australian
Vision Research (LAM), Perpetual IMPACT (LAM), and Fighting Blind-
ness Canada (LAM, ADP). ADP and LAM are Alcon Research Institute
Senior Investigators. ADP holds a Canada Research Chair (Tier 1). YS
was supported by postdoctoral fellowships from the Fonds de recherche
Sant´
e Qu´
ebec (FRQS) and CIHR, DVB was supported by a graduate
scholarship from FRQS, HQ was supported by a postdoctoral fellowship
from the Mexican National Council of Science and Technology
(CONACYT).
References
Abbott, N.J., 2002. Astrocyte-endothelial interactions and blood-brain barrier
permeability. J. Anatacosta 200, 629638.
Abran, D., Li, D.Y., Varma, D.R., Chemtob, S., 1995. Characterization and ontogeny of
PGE2 and PGF2 alpha receptors on the retinal vasculature of the pig. Prostaglandins
50, 253267.
Acosta, M.L., Mat Nor, M.N., Guo, C.X., Mugisho, O.O., Coutinho, F.P., Rupenthal, I.D.,
Green, C.R., 2021. Connexin therapeutics: blocking connexin hemichannel pores is
distinct from blocking pannexin channels or gap junctions. Neural Reg. Res. 16,
482488.
Adam, G., Schwartz, B., 1980. Increased uorescein lling defects in the wall of the optic
disc cup in glaucoma. Arch. Ophthalmol. 98, 15901592.
Agarwal, P., Agarwal, R., 2021. Tackling retinal ganglion cell apoptosis in glaucoma: role
of adenosine receptors. Expert Opin. Ther. Targets 25, 585596.
Ahmad, I., Subramani, M., 2022. Microglia: friends or foes in glaucoma? A
developmental perspective. Stem Cells Transl. Med. 11, 12101218.
Akimoto, S., Mitsumata, M., Sasaguri, T., Yoshida, Y., 2000. Laminar shear stress inhibits
vascular endothelial cell proliferation by inducing cyclin-dependent kinase inhibitor
p21(Sdi1/Cip1/Waf1). Circ. Res. 86, 185190.
Alarcon-Martinez, L., Shiga, Y., Villafranca-Baughman, D., Belforte, N., Quintero, H.,
Dotigny, F., Cueva Vargas, J., Di Polo, A., 2022. Pericyte dysfunction and loss of
inter-pericyte tunneling nanotubes promote neurovascular decits in glaucoma.
Proc. Natl. Acad. Sci. U. S. A. 119, e2110329119.
Alarcon-Martinez, L., Villafranca-Baughman, D., Quintero, H., Kacerovsky, J.B.,
Dotigny, F., Murai, K.K., Prat, A., Drapeau, P., Di Polo, A., 2020. Interpericyte
tunnelling nanotubes regulate neurovascular coupling. Nature 585, 9195.
Alarcon-Martinez, L., Yemisci, M., Dalkara, T., 2021. Pericyte morphology and function.
Histol. Histopathol. 36, 633643.
Alarcon-Martinez, L., Yilmaz-Ozcan, S., Yemisci, M., Schallek, J., Kılıç, K., Can, A., Di
Polo, A., Dalkara, T., 2018. Capillary pericytes express
α
-smooth muscle actin, which
requires prevention of lamentous-actin depolymerization for detection. Elife 7,
e34861.
Alarcon-Martinez, L., Yilmaz-Ozcan, S., Yemisci, M., Schallek, J., Kılıç, K., Villafranca-
Baughman, D., Can, A., Di Polo, A., Dalkara, T., 2019. Retinal ischemia induces
α
-SMA-mediated capillary pericyte contraction coincident with perivascular
glycogen depletion. Acta Neuropathol. Commun. 7, 134-134.
Allport, J.R., Muller, W.A., Luscinskas, F.W., 2000. Monocytes induce reversible focal
changes in vascular endothelial cadherin complex during transendothelial migration
under ow. J. Cell Biol. 148, 203216.
Allsopp, G., Gamble, H.J., 1979. An electron microscopic study of the pericytes of the
developing capillaries in human fetal brain and muscle. J. Anat. 128, 155168.
Alm, A., 1977. The effect of sympathetic stimulation on blood ow through t,e uvea,
retina and optic nerve in monkeys (Macacca irus). Exp. Eye Res. 25, 1924.
Almasieh, M., Wilson, A.M., Morquette, B., Cueva Vargas, J.L., Di Polo, A., 2012. The
molecular basis of retinal ganglion cell death in glaucoma. Prog. Retin. Eye Res. 31,
152181.
Anderson, D.R., Davis, E.B., 1996. Glaucoma, capillaries and pericytes. 5. Preliminary
evidence that carbon dioxide relaxes pericyte contractile tone. J. Int. dophtalmol.
210, 280284.
Ando, K., Tong, L., Peng, D., V´
azquez-Li´
ebanas, E., Chiyoda, H., He, L., Liu, J.,
Kawakami, K., Mochizuki, N., Fukuhara, S., Grutzendler, J., Betsholtz, C., 2022.
KCNJ8/ABCC9-containing K-ATP channel modulates brain vascular smooth muscle
development and neurovascular coupling. Dev. Cell 57, 13831399 e7.
Arai, H., Hori, S., Aramori, I., Ohkubo, H., Nakanishi, S., 1990. Cloning and expression of
a cDNA encoding an endothelin receptor. Nature 348, 730732.
Arango-Lievano, M., Boussadia, B., De Terdonck, L.D.T., Gault, C., Fontanaud, P.,
Lafont, C., Mollard, P., Marchi, N., Jeanneteau, F., 2018. Topographic reorganization
of cerebrovascular mural cells under seizure conditions. Cell Rep. 23, 10451059.
Arend, O., Plange, N., Sponsel, W.E., Remky, A., 2004. Pathogenetic aspects of the
glaucomatous optic neuropathy: uorescein angiographic ndings in patients with
primary open angle glaucoma. Brain Res. Bull. 62, 517524.
Arend, O., Remky, A., Plange, N., Kaup, M., Schwartz, B., 2005. Fluorescein leakage of
the optic disc in glaucomatous optic neuropathy. Graefes Arch. Clin. Exp.
Ophthalmol. 243, 659664.
Argaw, A.T., Gurfein, B.T., Zhang, Y., Zameer, A., John, G.R., 2009. VEGF-mediated
disruption of endothelial CLN-5 promotes blood-brain barrier breakdown. Proc. Natl.
Acad. Sci. U. S. A. 106, 19771982.
Armulik, A., Abramsson, A., Betsholtz, C., 2005. Endothelial/pericyte interactions. Circ.
Res. 97, 512523.
Armulik, A., Genov´
e, G., Betsholtz, C., 2011. Pericytes: developmental, physiological,
and pathological perspectives, problems, and promises. Dev. Cell 21, 193215.
Armulik, A., Genov´
e, G., M¨
ae, M., Nisancioglu, M.H., Wallgard, E., Niaudet, C., He, L.,
Norlin, J., Lindblom, P., Strittmatter, K., Johansson, B.R., Betsholtz, C., 2010.
Pericytes regulate the blood-brain barrier. Nature 468, 557561.
Arnold, A.C., 1995. Fluorescein angiographic characteristics of the optic disc in ischemic
and glaucomatous optic neuropathy. Curr. Opin. Ophthalmol. 6, 3035.
Arnold, T., Betsholtz, C., 2013. The importance of microglia in the development of the
vasculature in the central nervous system. Vasc. Cell 5, 4.
Ashworth, B., Rosen, E., 1970. Fluorescence of the optic disc in the late phase. Ann.
Ophthalmol. 1, 444451.
Attariwala, R., Giebs, C.P., Glucksberg, M.R., 1994. The inuence of elevated intraocular
pressure on vascular pressures in the cat retina. Invest. Ophthalmol. Vis. Sci. 35,
10191025.
Attwell, D., Buchan, A.M., Charpak, S., Lauritzen, M., Macvicar, B.A., Newman, E.A.,
2010. Glial and neuronal control of brain blood ow. Nature 468, 232243.
Attwell, D., Mishra, A., Hall, C.N., Ofarrell, F.M., Dalkara, T., 2016. What is a pericyte?
J. Cerebr. Blood Flow Metabol. 36, 451455.
Bandopadhyay, R., Orte, C., Lawrenson, J.G., Reid, A.R., De Silva, S., Allt, G., 2001.
Contractile proteins in pericytes at the blood-brain and blood-retinal barriers.
J. Neurocytol. 30, 3544.
Bao, L., Locovei, S., Dahl, G., 2004. Pannexin membrane channels are mechanosensitive
conduits for ATP. FEBS Lett. 572, 6568.
Barbe, M.T., Monyer, H., Bruzzone, R., 2006. Cell-cell communication beyond connexins:
the pannexin channels. Physiology 21, 103114.
Beckel, J.M., Argall, A.J., Lim, J.C., Xia, J., Lu, W., Coffey, E.E., Macarak, E.J.,
Shahidullah, M., Delamere, N.A., Zode, G.S., Shefeld, V.C., Shestopalov, V.I.,
Laties, A.M., Mitchell, C.H., 2014. Mechanosensitive release of adenosine 5-
triphosphate through pannexin channels and mechanosensitive upregulation of
pannexin channels in optic nerve head astrocytes: a mechanism for purinergic
involvement in chronic strain. Glia 62, 14861501.
Begg, I.S., Drance, S.M., Sweeney, V.P., 1970. Haemorrhage on the disc–a sign of acute
ischaemic optic neuropathy in chronic simple glaucoma. Can. J. Ophthalmol. 5,
321330.
Belforte, N., Agostinone, J., Alarcon-Martinez, L., Villafranca-Baughman, D., Dotigny, F.,
Cueva Vargas, J.L., Di Polo, A., 2021. AMPK hyperactivation promotes dendrite
retraction, synaptic loss, and neuronal dysfunction in glaucoma. Mol. Neurodegener.
16, 43.
Bell, R.D., Winkler, E.A., Sagare, A.P., Singh, I., Larue, B., Deane, R., Zlokovic, B.V.,
2010. Pericytes control key neurovascular functions and neuronal phenotype in the
adult brain and during brain aging. Neuron 68, 409427.
Benfey, N., Foubert, D., Ruthazer, E.S., 2022. Glia regulate the development, function,
and plasticity of the visual system from retina to cortex. Front. Neural Circ. 16,
826664.
Benigni, A., Cassis, P., Remuzzi, G., 2010. Angiotensin II revisited: new roles in
inammation, immunology and aging. EMBO Mol. Med. 2, 247257.
Bernstein, S.A., Fishman, G.I., 2016. Chapter 13 - connexins and heritable human
diseases. In: PITT, G.S. (Ed.), Ion Channels in Health and Disease. Academic Press,
Boston.
Berthiaume, A.A., Grant, R.I., Mcdowell, K.P., Underly, R.G., Hartmann, D.A., Levy, M.,
Bhat, N.R., Shih, A.Y., 2018. Dynamic remodeling of pericytes in vivo maintains
capillary coverage in the adult mouse brain. Cell Rep. 22, 816.
Beyer, E.C., Berthoud, V.M., 2018. Gap junction gene and protein families: connexins,
innexins, and pannexins. Biochim. Biophys. Acta Biomembr. 1860, 58.
Biesecker, K.R., Srienc, A.I., Shimoda, A.M., Agarwal, A., Bergles, D.E., Kofuji, P.,
Newman, E.A., 2016. Glial cell calcium signaling mediates capillary regulation of
blood ow in the retina. J. Neurosci. 36, 94359445.
Bill, A., 1975. Blood circulation and uid dynamics in the eye. Physiol. Rev. 55,
383417.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
17
Bill, A., 1985. Some aspects of the ocular circulation. Friedenwald lecture. Invest.
Ophthalmol. Vis. Sci. 26, 410424.
Bisht, K., Okojie, K.A., Sharma, K., Lentferink, D.H., Sun, Y.-Y., Chen, H.-R., Uweru, J.O.,
Amancherla, S., Calcuttawala, Z., Campos-Salazar, A.B., Corliss, B., Jabbour, L.,
Benderoth, J., Friestad, B., Mills, W.A., Isakson, B.E., Tremblay, M.-`
E., Kuan, C.-Y.,
Eyo, U.B., 2021. Capillary-associated microglia regulate vascular structure and
function through PANX1-P2RY12 coupling in mice. Nat. Commun. 12, 5289.
Blanco, R., Martínez-Navarrete, G., Valiente-Soriano, F.J., Avil´
es-Trigueros, M., P´
erez-
Rico, C., Serrano-Puebla, A., Boya, P., Fern´
andez, E., Vidal-Sanz, M., De La Villa, P.,
2017. The S1P1 receptor-selective agonist CYM-5442 protects retinal ganglion cells
in endothelin-1 induced retinal ganglion cell loss. Exp. Eye Res. 164, 3745.
Blanco, V.M., Stern, J.E., Filosa, J.A., 2008. Tone-dependent vascular responses to
astrocyte-derived signals. Am. J. Physiol. Heart Circ. Physiol. 294, H2855H2863.
Boal, A.M., Risner, M.L., Cooper, M.L., Wareham, L.K., Calkins, D.J., 2021. Astrocyte
networks as therapeutic targets in glaucomatous neurodegeneration. Cells 10.
Bohannon, D.G., Long, D., Kim, W.K., 2021. Understanding the heterogeneity of human
pericyte subsets in blood-brain barrier homeostasis and neurological diseases. Cells
10, 890.
Boia, R., Salinas-Navarro, M., Gallego-Ortega, A., Galindo-Romero, C., Aires, I.D., Agudo-
Barriuso, M., Ambrosio, A.F., Vidal-Sanz, M., Santiago, A.R., 2020. Activation of
adenosine A3 receptor protects retinal ganglion cells from degeneration induced by
ocular hypertension. Cell Death Dis. 11, 401.
Bonomi, L., Marchini, G., Marraffa, M., Bernardi, P., Morbio, R., Varotto, A., 2000.
Vascular risk factors for primary open angle glaucoma: the Egna-Neumarkt Study.
Ophthalmology 107, 12871293.
Borysova, L., Wray, S., Eisner, D.A., Burdyga, T., 2013. How calcium signals in myocytes
and pericytes are integrated across in situ microvascular networks and control
microvascular tone. Cell Calcium 54, 163174.
Bradley, K.K., Jaggar, J.H., Bonev, A.D., Heppner, T.J., Flynn, E.R., Nelson, M.T.,
Horowitz, B., 1999. Kir2.1 encodes the inward rectier potassium channel in rat
arterial smooth muscle cells. J. Physiol. 515 (Pt 3), 639651.
Brasko, C., Butt, A.M., 2018. Expression of Kir2.1 inward rectifying potassium channels
in optic nerve glia: evidence for heteromeric association with Kir4.1 and Kir5.1.
Neuroglia 1, 176187.
Brockhaus, K., Melkonyan, H., Prokosch-Willing, V., Liu, H., Thanos, S., 2020.
Alterations in tight- and adherens-junction proteins related to glaucoma mimicked in
the organotypically cultivated mouse retina under elevated pressure. Invest.
Ophthalmol. Vis. Sci. 61, 46.
Budenz, D.L., Huecker, J.B., Gedde, S.J., Gordon, M., Kass, M., 2017. Thirteen-year
follow-up of optic disc hemorrhages in the ocular hypertension treatment study. Am.
J. Ophthalmol. 174, 126133.
Buerk, D.G., Riva, C.E., 2002. Adenosine enhances functional activation of blood ow in
cat optic nerve head during photic stimulation independently from nitric oxide.
Microvasc. Res. 64, 254264.
Burdyga, T., Borysova, L., 2018. Ca(2+) signalling in pericytes. Adv. Exp. Med. Biol.
1109, 95109.
Burgoyne, C.F., 2011. A biomechanical paradigm for axonal insult within the optic nerve
head in aging and glaucoma. Exp. Eye Res. 93, 120132.
Burgoyne, C.F., Downs, J.C., Bellezza, A.J., Suh, J.K.F., Hart, R.T., 2005. The optic nerve
head as a biomechanical structure: a new paradigm for understanding the role of
IOP-related stress and strain in the pathophysiology of glaucomatous optic nerve
head damage. Prog. Retin. Eye Res. 24, 3973.
Calder, B.W., Matthew Rhett, J., Bainbridge, H., Fann, S.A., Gourdie, R.G., Yost, M.J.,
2015. Inhibition of connexin 43 hemichannel-mediated ATP release attenuates early
inammation during the foreign body response. Tissue Eng. 21, 17521762.
Calkins, D.J., 2012. Critical pathogenic events underlying progression of
neurodegeneration in glaucoma. Prog. Retin. Eye Res. 31, 702719.
Campagno, K.E., Lu, W., Jassim, A.H., Albalawi, F., Cenaj, A., Tso, H.Y., Clark, S.P.,
Sripinun, P., G´
omez, N.M., Mitchell, C.H., 2021. Rapid morphologic changes to
microglial cells and upregulation of mixed microglial activation state markers
induced by P2X7 receptor stimulation and increased intraocular pressure.
J. Neuroinammation 18, 217.
Campbell, W.B., Fleming, I., 2010. Epoxyeicosatrienoic acids and endothelium-
dependent responses. Pügers Arch. 459, 881895.
Carelli, V., La Morgia, C., Valentino, M.L., Barboni, P., Ross-Cisneros, F.N., Sadun, A.A.,
2009. Retinal ganglion cell neurodegeneration in mitochondrial inherited disorders.
Biochim. Biophys. Acta 1787, 518528.
Casson, R.J., Chidlow, G., Crowston, J.G., Williams, P.A., Wood, J.P.M., 2020. Retinal
energy metabolism in health and glaucoma. Prog. Retin. Eye Res., 100881
Chan-Ling, T., Koina, M.E., Mccolm, J.R., Dahlstrom, J.E., Bean, E., Adamson, S., Yun, S.,
Baxter, L., 2011. Role of CD44+stem cells in mural cell formation in the human
choroid: evidence of vascular instability due to limited pericyte ensheathment.
Invest. Ophthalmol. Vis. Sci. 52, 399410.
Chauhan, B., 2008. Endothelin and its potential role in glaucoma. Can. J. Ophthalmol.
43, 356360.
Chauhan, B.C., Levatte, T.L., Jollimore, C.A., Yu, P.K., Reitsamer, H.A., Kelly, M.E.M.,
Yu, D.-Y., Tremblay, F.O., Archibald, M.L., 2004. Model of endothelin-1-induced
chronic optic neuropathy in rat. Invest. Ophthalmol. Vis. Sci. 45, 144152.
Checchin, D., Sennlaub, F., Levavasseur, E., Leduc, M., Chemtob, S., 2006. Potential role
of microglia in retinal blood vessel formation. Invest. Ophthalmol. Vis. Sci. 47,
35953602.
Chen, S., Tisch, N., Kegel, M., Yerbes, R., Hermann, R., Hudalla, H., Zuliani, C.,
Gulculer, G.S., Zwadlo, K., Von Engelhardt, J., Ruiz De Almodovar, C., Martin-
Villalba, A., 2017. CNS macrophages control neurovascular development via CD95L.
Cell Rep. 19, 13781393.
Chen, Y.-S., Green, C.R., Wang, K., Danesh-Meyer, H.V., Rupenthal, I.D., 2015. Sustained
intravitreal delivery of connexin43 mimetic peptide by poly(D,L-lactide-co-
glycolide) acid micro- and nanoparticles–Closing the gap in retinal ischaemia. Eur. J.
Pharm. Biopharm. 95, 378386.
Chen, Y., Mccarron, R.M., Azzam, N., Bembry, J., Reutzler, C., Lenz, F.A., Spatz, M.,
2000. Endothelin-1 and nitric oxide affect human cerebromicrovascular endothelial
responses and signal transduction. Acta Neurochir. 76, 131135.
Cherecheanu, A.P., Garhofer, G., Schmidl, D., Werkmeister, R., Schmetterer, L., 2012.
Ocular perfusion pressure and ocular blood ow in glaucoma. Curr. Opin.
Pharmacol. 13, 17.
Chew, S.S., Johnson, C.S., Green, C.R., Danesh-Meyer, H.V., 2011. Response of retinal
Connexin43 to optic nerve injury. Invest. Ophthalmol. Vis. Sci. 52, 36203629.
Choi, J., Kim, K.H., Jeong, J., Cho, H.S., Lee, C.H., Kook, M.S., 2007. Circadian
uctuation of mean ocular perfusion pressure is a consistent risk factor for normal-
tension glaucoma. Invest. Ophthalmol. Vis. Sci. 48, 104111.
Chuquet, J., Hollender, L., Nimchinsky, E.A., 2007. High-resolution in vivo imaging of
the neurovascular unit during spreading depression. J. Neurosci. 27, 40364044.
Ciof Ga, V.B.E., 1996. Vasculature of the anterior optic nerve and peripapillary choroid.
In: RITCH R, S.M., KRUPIN, T. (Eds.), The Glaucomas: Glaucoma Therapy.
Contreras, J.E., S´
aez, J.C., Bukauskas, F.F., Bennett, M.V.L., 2003. Gating and regulation
of connexin 43 (Cx43) hemichannels. Proc. Natl. Acad. Sci. U. S. A. 100,
1138811393.
Costa, V.P., Harris, A., Anderson, D., Stodtmeister, R., Cremasco, F., Kergoat, H.,
Lovasik, J., Stalmans, I., Zeitz, O., Lanzl, I., Gugleta, K., Schmetterer, L., 2014.
Ocular perfusion pressure in glaucoma. Acta Ophthalmol. 92, e252e266.
Cotrina, M.L., Lin, J.H., Alves-Rodrigues, A., Liu, S., Li, J., Azmi-Ghadimi, H., Kang, J.,
Naus, C.C., Nedergaard, M., 1998. Connexins regulate calcium signaling by
controlling ATP release. Proc. Natl. Acad. Sci. U. S. A. 95, 1573515740.
Cotrina, M.L., Nedergaard, M., 2009. Physiological and pathological functions of P2X7
receptor in the spinal cord. Purinergic Signal. 5, 223232.
Crish, S.D., Sappington, R.M., Inman, D.M., Horner, P.J., Calkins, D.J., 2010. Distal
axonopathy with structural persistence in glaucomatous neurodegeneration. Proc.
Natl. Acad. Sci. U. S. A. 107, 51965201.
Cs´
asz´
ar, E., L´
en´
art, N., Cser´
ep, C., K¨
ornyei, Z., Fekete, R., P´
osfai, B., Bal´
azs, D.,
Hangya, B., Schwarcz, A.D., Szabadits, E., Sz¨
oll˝
osi, D., Szigeti, K., M´
ath´
e, D., West, B.
L., Sviatk´
o, K., Br´
as, A.R., Mariani, J.-C., Kliewer, A., Lenkei, Z., Hricis´
ak, L.,
Beny´
o, Z., Baranyi, M., Sperl´
agh, B., Menyh´
art, ´
A., Farkas, E., D´
enes, ´
A., 2022.
Microglia modulate blood ow, neurovascular coupling, and hypoperfusion via
purinergic actions. J. Exp. Med. 219, e20211071.
Csaszar, E., Lenart, N., Cserep, C., Kornyei, Z., Fekete, R., Posfai, B., Balazs, D.,
Hangya, B., Schwarcz, A.D., Szabadits, E., Szollosi, D., Szigeti, K., Mathe, D., West, B.
L., Sviatko, K., Bras, A.R., Mariani, J.C., Kliewer, A., Lenkei, Z., Hricisak, L.,
Benyo, Z., Baranyi, M., Sperlagh, B., Menyhart, A., Farkas, E., Denes, A., 2022.
Microglia modulate blood ow, neurovascular coupling, and hypoperfusion via
purinergic actions. J. Exp. Med. 219.
Cueva Vargas, J.L., Belforte, N., Di Polo, A., 2016. The glial cell modulator ibudilast
attenuates neuroinammation and enhances retinal ganglion cell viability in
glaucoma through protein kinase A signaling. Neurobiol. Dis. 93, 156171.
Cueva Vargas, J.L., Osswald, I.K., Unsain, N., Aurousseau, M.R., Barker, P.A., Bowie, D.,
Di Polo, A., 2015. Soluble tumor necrosis factor alpha promotes retinal ganglion cell
death in glaucoma via calcium-permeable AMPA receptor activation. J. Neurosci.
35, 1208812102.
Cuevas, P., Gutierrez-Diaz, J.A., Reimers, D., Dujovny, M., Diaz, F.G., Ausman, J.I., 1984.
Pericyte endothelial gap junctions in human cerebral capillaries. Anat. Embryol.
170, 155159.
Cunha-Vaz, J.G., 2004. The blood-retinal barriers system. Basic concepts and clinical
evaluation. Exp. Eye Res. 78, 715721.
Dabertrand, F., Harraz, O.F., Koide, M., Longden, T.A., Rosehart, A.C., Hill-Eubanks, D.
C., Joutel, A., Nelson, M.T., 2021. PIP(2) corrects cerebral blood ow decits in
small vessel disease by rescuing capillary Kir2.1 activity. Proc. Natl. Acad. Sci. U. S.
A. 118.
Dalkara, T., Alarcon-Martinez, L., 2015. Cerebral microvascular pericytes and
neurogliovascular signaling in health and disease. Brain Res. 1623, 317.
Daneman, R., Prat, A., 2015. The blood-brain barrier. Cold Spring Harbor Perspect. Biol.
7 a020412-a020412.
Daneman, R., Zhou, L., Kebede, A.A., Barres, B.A., 2010. Pericytes are required for
bloodbrain barrier integrity during embryogenesis. Nature 468, 562566.
Danesh-Meyer, H.V., Huang, R., Nicholson, L.F.B., Green, C.R., 2008. Connexin43
antisense oligodeoxynucleotide treatment down-regulates the inammatory
response in an in vitro interphase organotypic culture model of optic nerve
ischaemia. J. Clin. Neurosci. 15, 12531263.
Danesh-Meyer, H.V., Kerr, N.M., Zhang, J., Eady, E.K., Ocarroll, S.J., Nicholson, L.F.,
Johnson, C.S., Green, C.R., 2012. Connexin43 mimetic peptide reduces vascular leak
and retinal ganglion cell death following retinal ischaemia. Brain 135, 506520.
Danesh-Meyer, H.V., Zhang, J., Acosta, M.L., Rupenthal, I.D., Green, C.R., 2016.
Connexin43 in retinal injury and disease. Prog. Retin. Eye Res. 51, 4168.
Davidge, S.T., Baker, P.N., Laughlin, M.K., Roberts, J.M., 1995. Nitric oxide produced by
endothelial cells increases production of eicosanoids through activation of
prostaglandin H synthase. Circ. Res. 77, 274283.
Davidson, J.O., Green, C.R., Bennet, L., Nicholson, L.F.B., Danesh-Meyer, H., Ocarroll, S.
J., Gunn, A.J., 2013. A key role for connexin hemichannels in spreading ischemic
brain injury. Curr. Drug Targets 14, 3646.
Davis, M.J., Hill, M.A., 1999. Signaling mechanisms underlying the vascular myogenic
response. Physiol. Rev. 79, 387423.
Dejana, E., Vestweber, D., 2013. The role of VE-cadherin in vascular morphogenesis and
permeability control. Prog. Mol. Biol. Transl. Sci. 116, 119144.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
18
Di Polo, A., Aigner, L.J., Dunn, R.J., Bray, G.M., Aguayo, A.J., 1998. Prolonged delivery
of brain-derived neurotrophic factor by adenovirus-infected Müller cells temporarily
rescues injured retinal ganglion cells. Proc. Natl. Acad. Sci. U. S. A. 95, 39783983.
Donati, G., Pournaras, C.J., Munoz, J.L., Poitry, S., Poitry-Yamate, C.L., Tsacopoulos, M.,
1995. Nitric oxide controls arteriolar tone in the retina of the miniature pig. Invest.
Ophthalmol. Vis. Sci. 36, 22282237.
Dorner, G.T., Garhofer, G., Kiss, B., Polska, E., Polak, K., Riva, C.E., Schmetterer, L.,
2003. Nitric oxide regulates retinal vascular tone in humans. Am. J. Physiol. 285,
H631H636.
Downs, J.C., 2015. Optic nerve head biomechanics in aging and disease. Exp. Eye Res.
133, 1929.
Drance, S.M., 1989. Disc hemorrhages in the glaucomas. Surv. Ophthalmol. 33, 331337.
Drance, S.M., Begg, I.S., 1970. Sector haemorrhage–a probable acute ischaemic disc
change in chronic simple glaucoma. Can. J. Ophthalmol. 5, 137141.
Duan, A., Bedggood, P.A., Bui, B.V., Metha, A.B., 2016. Evidence of icker-induced
functional hyperaemia in the smallest vessels of the human retinal blood supply.
PLoS One 11, e0162621.
Dubyak, G.R., 2009. Both sides now: multiple interactions of ATP with pannexin-1
hemichannels. Focus on "A permeant regulating its permeation pore: inhibition of
pannexin 1 channels by ATP". Am. J. Physiol. 296, C235C241.
Eberth, C., 1871. Handbuch der Lehre von der Gewegen des Menschen und der Tiere.
Engelmann, Leipzig, Ger.
Emmerich, A., 1876. Über Glaucoma Haemorrhagicum (Berlin).
Enge, M., 2002. Endothelium-specic platelet-derived growth factor-B ablation mimics
diabetic retinopathy. EMBO J. 21, 43074316.
Eperon, G., Johnson, M., David, N.J., 1975. The effect of arterial PO2 on relative retinal
blood ow in monkeys. Invest. Ophthalmol. 14, 342352.
Fallon, T.J., Maxwell, D., Kohner, E.M., 1985. Retinal vascular autoregulation in
conditions of hyperoxia and hypoxia using the blue eld entoptic phenomenon.
Ophthalmology 92, 701705.
Fan, L.M., Douglas, G., Bendall, J.K., Mcneill, E., Crabtree, M.J., Hale, A.B., Mai, A., Li, J.
M., Mcateer, M.A., Schneider, J.E., Choudhury, R.P., Channon, K.M., 2014.
Endothelial cell-specic reactive oxygen species production increases susceptibility
to aortic dissection. Circulation 129, 26612672.
Feke, G.T., Pasquale, L.R., 2008. Retinal blood ow response to posture change in
glaucoma patients compared with healthy subjects. Ophthalmology 115, 246252.
F´
el´
etou, M., Vanhoutte, P.M., 2006. Endothelium-derived hyperpolarizing factor: where
are we now? Arterioscler. Thromb. Vasc. Biol. 26, 12151225.
Fishbein, S.L., Schwartz, B., 1977. Optic disc in glaucoma: topography and extent of
uorescein lling defects. Arch. Ophthalmol. 95, 19751979.
Flammer, J., Haeiger, I.O., Orgül, S., Resink, T., 1999. Vascular dysregulation: a
principal risk factor for glaucomatous damage? J. Glaucoma 8, 212219.
Flammer, J., Orgül, S., Costa, V.P., Orzalesi, N., Krieglstein, G.K., Serra, L.M., Renard, J.
P., Stef´
ansson, E., 2002. The impact of ocular blood ow in glaucoma. Prog. Retin.
Eye Res. 21, 359393.
Flammer, J., Pache, M., Resink, T., 2001. Vasospasm, its role in the pathogenesis of
diseases with particular reference to the eye. Prog. Retin. Eye Res. 20, 319349.
Foulquier, S., Caolo, V., Swennen, G., Milanova, I., Reinhold, S., Recarti, C., Alenina, N.,
Bader, M., Steckelings, U.M., Vanmierlo, T., Post, M.J., Jones, E.A., Van
Oostenbrugge, R.J., Unger, T., 2019. The role of receptor MAS in microglia-driven
retinal vascular development. Angiogenesis 22, 481489.
Franco, C.A., Jones, M.L., Bernabeu, M.O., Geudens, I., Mathivet, T., Rosa, A., Lopes, F.
M., Lima, A.P., Ragab, A., Collins, R.T., Phng, L.-K., Coveney, P.V., Gerhardt, H.,
2015. Dynamic endothelial cell rearrangements drive developmental vessel
regression. PLoS Biol. 13, e1002125-e1002125.
Freeman, R.D., Li, B., 2016. Neural-metabolic coupling in the central visual pathway.
Philos. Trans. R. Soc. Lond. B Biol. Sci. 371.
Fresta, C.G., Fidilio, A., Caruso, G., Caraci, F., Giblin, F.J., Marco Leggio, G.,
Salomone, S., Drago, F., Bucolo, C., 2020. A new human bloodretinal barrier model
based on endothelial cells, pericytes, and astrocytes. Int. J. Mol. Sci. 21, 1636.
Friedland, R.P., Iadecola, C., 1991. Roy and Sherrington (1890): a centennial
reexamination of "On the regulation of the blood-supply of the brain". Neurology 41,
1014.
Fry, L.E., Fahy, E., Chrysostomou, V., Hui, F., Tang, J., Van Wijngaarden, P., Petrou, S.,
Crowston, J.G., 2018. The coma in glaucoma: retinal ganglion cell dysfunction and
recovery. Prog. Retin. Eye Res. 65, 7792.
Galinsky, R., Davidson, J.O., Dean, J.M., Green, C.R., Bennet, L., Gunn, A.J., 2018. Glia
and hemichannels: key mediators of perinatal encephalopathy. Neural Reg. Res. 13,
181189.
Gardiner, S.K., Cull, G., Fortune, B., 2023. Retinal vessel pulsatile characteristics
associated with vascular stiffness can predict the rate of functional progression in
glaucoma suspects. Invest. Ophthalmol. Vis. Sci. 64, 30.
Garh¨
ofer, G., Chua, J., Tan, B., Wong, D., Schmidl, D., Schmetterer, L., 2020. Retinal
neurovascular coupling in diabetes. J. Clin. Med. 9.
Garh¨
ofer, G., Resch, H., Weigert, G., Lung, S., Simader, C., Schmetterer, L., 2005. Short-
term increase of intraocular pressure does not alter the response of retinal and optic
nerve head blood ow to icker stimulation. Invest. Ophthalmol. Vis. Sci. 46,
17211725.
Garh¨
ofer, G., Zawinka, C., Huemer, K.-H., Schmetterer, L., Dorner, G.T., 2003. Flicker
light-induced vasodilatation in the human retina: effect of lactate and changes in
mean arterial pressure. Invest. Ophthalmol. Vis. Sci. 44, 53095314.
Garh¨
ofer, G., Zawinka, C., Resch, H., Huemer, K., Schmetterer, L., Dorner, G., 2004.
Response of retinal vessel diameters to icker stimulation in patients with early open
angle glaucoma. J. Glaucoma 13, 340344.
Garrison, A.T., Bignold, R.E., Wu, X., Johnson, J.R., 2023. Pericytes: the lung-forgotten
cell type. Front. Physiol. 14, 1150028.
Gauthier, R., Joly, S., Pernet, V., Lachapelle, P., Di Polo, A., 2005. Brain-derived
neurotrophic factor gene delivery to Müller glia preserves structure and function of
light-damaged photoreceptors. Invest. Ophthalmol. Vis. Sci. 46, 33833392.
Geevarghese, A., Herman, I.M., 2014. Pericyte-endothelial crosstalk: implications and
opportunities for advanced cellular therapies. Transl. Res. 163, 296306.
Geijer, C., Bill, A., 1979. Effects of raised intraocular pressure on retinal, prelaminar,
laminar, and retrolaminar optic nerve blood ow in monkeys. Invest. Ophthalmol.
Vis. Sci. 18, 10301042.
Geyman, L.S., Garg, R.A., Suwan, Y., Trivedi, V., Krawitz, B.D., Mo, S., Pinhas, A.,
Tantraworasin, A., Chui, T.Y.P., Ritch, R., Rosen, R.B., 2017. Peripapillary perfused
capillary density in primary open-angle glaucoma across disease stage: an optical
coherence tomography angiography study. Br. J. Ophthalmol. 101, 12611268.
Gherghel, D., Hosking, S.L., Cunliffe, I.A., 2004. Abnormal systemic and ocular vascular
response to temperature provocation in primary open-angle glaucoma patients: a
case for autonomic failure? Invest. Ophthalmol. Vis. Sci. 45, 35463554.
Giaume, C., Leybaert, L., Naus, C.C., S´
aez, J.C., 2013. Connexin and pannexin
hemichannels in brain glial cells: properties, pharmacology, and roles. Front.
Pharmacol. 4, 88.
Giri, R., Shen, Y., Stins, M., Du Yan, S., Schmidt, A.M., Stern, D., Kim, K.S., Zlokovic, B.,
Kalra, V.K., 2000. beta-amyloid-induced migration of monocytes across human brain
endothelial cells involves RAGE and PECAM-1. Am. J. Physiol. Cell Physiol. 279,
C1772C1781.
Glucksberg, M.R., Dunn, R., 1993. Direct measurement of retinal microvascular pressures
in the live, anesthetized cat. Microvasc. Res. 45, 158165.
Gonzales, A.L., Klug, N.R., Moshkforoush, A., Lee, J.C., Lee, F.K., Shui, B., Tsoukias, N.
M., Kotlikoff, M.I., Hill-Eubanks, D., Nelson, M.T., 2020. Contractile pericytes
determine the direction of blood ow at capillary junctions. Proc. Natl. Acad. Sci. U.
S. A. 117, 2702227033.
Gordon, G.R.J., Choi, H.B., Rungta, R.L., Ellis-Davies, G.C.R., Macvicar, B.A., 2008. Brain
metabolism dictates the polarity of astrocyte control over arterioles. Nature 456,
745749.
Grant, R.I., Hartmann, D.A., Underly, R.G., Berthiaume, A.A., Bhat, N.R., Shih, A.Y.,
2019. Organizational hierarchy and structural diversity of microvascular pericytes in
adult mouse cortex. J. Cerebr. Blood Flow Metabol. 39, 411425.
Greene, C., Hanley, N., Campbell, M., 2019. Claudin-5: gatekeeper of neurological
function. Fluids Barriers CNS 16, 3.
Grieshaber, M.C., Mozaffarieh, M., Flammer, J., 2007. What is the link between vascular
dysregulation and glaucoma? Surv. Ohthalmol. 52, S144S154.
Grieshaber, M.C., Terhorst, T., Flammer, J., 2006. The pathogenesis of optic disc splinter
haemorrhages: a new hypothesis. Acta Ophthalmol. Scand. 84, 6268.
Grunwald, J., Riva, C., Stone, R., Keates, E., Petrig, B., 1984a. Retinal autoregulation in
open-angle glaucoma. Ophthalmol. Times 91, 16901694.
Grunwald, J.E., Riva, C.E., Stone, R.A., Keates, E.U., Petrig, B.L., 1984b. Retinal
autoregulation in open-angle glaucoma. Ophthalmol. Times 91, 16901694.
Guedes, G., Tsai, J.C., Loewen, N.A., 2011. Glaucoma and aging. Curr. Aging Sci. 4,
110117.
Gugleta, K., Fuchsj¨
ager-Mayrl, G., Orgül, S., 2007. Is neurovascular coupling of relevance
in glaucoma? Surv. Ophthalmol. 52 (Suppl. 2), S139S143.
Gugleta, K., Kochkorov, A., Waldmann, N., Polunina, A., Katamay, R., Flammer, J.,
Orgul, S., 2012. Dynamics of retinal vessel response to icker light in glaucoma
patients and ocular hypertensives. Graefes Arch. Clin. Exp. Ophthalmol. 250,
589594.
Gugleta, K., Türksever, C., Polunina, A., Orgül, S., 2013. Effect of ageing on the retinal
vascular responsiveness to icker light in glaucoma patients and in ocular
hypertension. Br. J. Ophthalmol. 97, 848851.
Gugleta, K., Zawinka, C., Rickenbacher, I., Kochkorov, A., Katamay, R., Flammer, J.,
Orgul, S., 2006. Analysis of retinal vasodilation after icker light stimulation in
relation to vasospastic propensity. Invest. Ophthalmol. Vis. Sci. 47, 40344041.
Guidoboni, G., Harris, A., Cassani, S., Arciero, J., Siesky, B., Amireskandari, A., Tobe, L.,
Egan, P., Januleviciene, I., Park, J., 2014. Intraocular pressure, blood pressure, and
retinal blood ow autoregulation: a mathematical model to clarify their relationship
and clinical relevance. Invest. Ophthalmol. Vis. Sci. 55, 41054118.
Guo, C.X., Mat Nor, M.N., Danesh-Meyer, H.V., Vessey, K.A., Fletcher, E.L., Ocarroll, S.
J., Acosta, M.L., Green, C.R., 2016. Connexin43 mimetic peptide improves retinal
function and reduces inammation in a light-damaged albino rat model. Invest.
Ophthalmol. Vis. Sci. 57, 39613973.
Guo, L., Moss, S.E., Alexander, R.A., Ali, R.R., Fitzke, F.W., Cordeiro, M.F., 2005. Retinal
ganglion cell apoptosis in glaucoma is related to intraocular pressure and IOP-
induced effects on extracellular matrix. Invest. Ophthalmol. Vis. Sci. 46, 175.
Haider, A.A., Rex, T.S., Wareham, L.K., 2022. cGMP signaling in the neurovascular unit-
implications for retinal ganglion cell survival in glaucoma. Biomolecules 12.
Hall, C.N., Reynell, C., Gesslein, B., Hamilton, N.B., Mishra, A., Sutherland, B.A.,
Ofarrell, F.M., Buchan, A.M., Lauritzen, M., Attwell, D., 2014. Capillary pericytes
regulate cerebral blood ow in health and disease. Nature 508, 5560.
Hamilton, N.B., Attwell, D., Hall, C.N., 2010. Pericyte-mediated regulation of capillary
diameter: a component of neurovascular coupling in health and disease. Front.
Neuroenergetics 2, 5.
Hammond, T.R., Robinton, D., Stevens, B., 2018. Microglia and the brain:
complementary partners in development and disease. Annu. Rev. Cell Dev. Biol. 34,
523544.
Hariharan, A., Robertson, C.D., Garcia, D.C.G., Longden, T.A., 2022. Brain capillary
pericytes are metabolic sentinels that control blood ow through a K(ATP) channel-
dependent energy switch. Cell Rep. 41, 111872.
Hariharan, A., Weir, N., Robertson, C., He, L., Betsholtz, C., Longden, T.A., 2020. The ion
channel and GPCR toolkit of brain capillary pericytes. Front. Cell. Neurosci. 14,
601324.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
19
Harraz, O.F., Longden, T.A., Dabertrand, F., Hill-Eubanks, D., Nelson, M.T., 2018a.
Endothelial GqPCR activity controls capillary electrical signaling and brain blood
ow through PIP2 depletion. Proc. Natl. Acad. Sci. U. S. A. 115, E3569E3577.
Harraz, O.F., Longden, T.A., Hill-Eubanks, D., Nelson, M.T., 2018b. PIP2 depletion
promotes TRPV4 channel activity in mouse brain capillary endothelial cells. Elife 7,
e38689.
Harris, A., Ciulla, T.A., Chung, H.S., Martin, B., 1998. Regulation of retinal and optic
nerve blood ow. Arch. Ophthalmol. 116, 14911495.
Harris, F.S., Rhoton, A.L., 1976. Anatomy of the cavernous sinus. A microsurgical study.
J. Neurosurg. 45, 169180.
Hartmann, D.A., Berthiaume, A.-A., Grant, R.I., Harrill, S.A., Koski, T., Tieu, T.,
Mcdowell, K.P., Faino, A.V., Kelly, A.L., Shih, A.Y., 2021. Brain capillary pericytes
exert a substantial but slow inuence on blood ow. Nat. Neurosci. 24, 633645.
Hartmann, D.A., Coelho-Santos, V., Shih, A.Y., 2022. Pericyte control of blood ow
across microvascular zones in the central nervous system. Annu. Rev. Physiol. 84,
331354.
Hattori, Y., Itoh, H., Tsugawa, Y., Nishida, Y., Kurata, K., Uemura, A., Miyata, T., 2022.
Embryonic pericytes promote microglial homeostasis and their effects on neural
progenitors in the developing cerebral cortex. J. Neurosci. 42, 362376.
Haynes, S.E., Hollopeter, G., Yang, G., Kurpius, D., Dailey, M.E., Gan, W.B., Julius, D.,
2006. The P2Y12 receptor regulates microglial activation by extracellular
nucleotides. Nat. Neurosci. 9, 15121519.
Hayreh, S.S., 1997. Anterior ischemic optic neuropathy. Clin. Neurosci. 4, 251263.
Hayreh, S.S., 2001. Blood ow in the optic nerve head and factors that may inuence it.
Prog. Retin. Eye Res. 20, 595624.
He, L., Vanlandewijck, M., M¨
ae, M.A., Andrae, J., Ando, K., Del Gaudio, F., Nahar, K.,
Lebouvier, T., Lavi˜
na, B., Gouveia, L., Sun, Y., Raschperger, E., Segerstolpe, Å.,
Liu, J., Gustafsson, S., R¨
as¨
anen, M., Zarb, Y., Mochizuki, N., Keller, A., Lendahl, U.,
Betsholtz, C., 2018. Single-cell RNA sequencing of mouse brain and lung vascular
and vessel-associated cell types. Sci. Data 5, 180160.
He, L., Vanlandewijck, M., Raschperger, E., Andaloussi M¨
ae, M., Jung, B., Lebouvier, T.,
Ando, K., Hofmann, J., Keller, A., Betsholtz, C., 2016. Analysis of the brain mural cell
transcriptome. Sci. Rep. 6, 35108.
Henkind, P., 1967. Radial peripapillary capillaries of the retina. I. Anatomy: human and
comparative. Br. J. Ophthalmol. 51, 115123.
Henry, E., Newby, D.E., Webb, D.J., Obrien, C., 1999. Peripheral endothelial
dysfunction in normal pressure glaucoma. Invest. Ophthalmol. Vis. Sci. 40,
17101714.
Hickam, J.B., Frayser, R., 1966. Studies of the retinal circulation in man. Observations on
vessel diameter, arteriovenous oxygen difference, and mean circulation time.
Circulation 33, 302316.
Hill, R.A., Tong, L., Yuan, P., Murikinati, S., Gupta, S., Grutzendler, J., 2015. Regional
blood ow in the normal and ischemic brain is controlled by arteriolar smooth
muscle cell contractility and not by capillary pericytes. Neuron 87, 95110.
Hirano, T., Chanwimol, K., Weichsel, J., Tepelus, T., Sadda, S., 2018. Distinct retinal
capillary plexuses in normal eyes as observed in optical coherence tomography
angiography axial prole analysis. Sci. Rep. 8, 9380.
Hirunpattarasilp, C., James, G., Kwanthongdee, J., Freitas, F., Huo, J., Sethi, H.,
Kittler, J.T., Owens, R.J., Mccoy, L.E., Attwell, D., 2023. SARS-CoV-2 triggers
pericyte-mediated cerebral capillary constriction. Brain 146, 727738.
Hogan, M.J., Feeney, L., 1963. The ultrastructure of retinal vessels. II. The small vessels.
maynard. J Ultrastruct Res 49, 2946.
Hoiland, R.L., Fisher, J.A., Ainslie, P.N., 2019. Regulation of the cerebral circulation by
arterial carbon dioxide. Compr. Physiol. 9, 11011154.
Hørlyck, S., Cai, C., Helms, H.C.C., Lauritzen, M., Brodin, B., 2021. ATP induces
contraction of cultured brain capillary pericytes via activation of P2Y-type
purinergic receptors. Am. J. Physiol. Heart Circ. Physiol. 320, H699h712.
Hormel, T.T., Jia, Y., Jian, Y., Hwang, T.S., Bailey, S.T., Pennesi, M.E., Wilson, D.J.,
Morrison, J.C., Huang, D., 2021. Plexus-specic retinal vascular anatomy and
pathologies as seen by projection-resolved optical coherence tomographic
angiography. Prog. Retin. Eye Res. 80, 100878.
Hosford, P.S., Gourine, A.V., 2019. What is the key mediator of the neurovascular
coupling response? Neurosci. Biobehav. Rev. 96, 174181.
Howell, G.R., Macalinao, D.G., Sousa, G.L., Walden, M., Soto, I., Kneeland, S.C.,
Barbay, J.M., King, B.L., Marchant, J.K., Hibbs, M., Stevens, B., Barres, B.A.,
Clark, A.F., Libby, R.T., John, S.W.M., 2011. Molecular clustering identies
complement and endothelin induction as early events in a mouse model of glaucoma.
J. Clin. Invest. 121, 14291444.
Howell, G.R., Macnicoll, K.H., Braine, C.E., Soto, I., Macalinao, D.G., Sousa, G.L., John, S.
W., 2014. Combinatorial targeting of early pathways profoundly inhibits
neurodegeneration in a mouse model of glaucoma. Neurobiol. Dis. 71, 4452.
Howell, G.R., Soto, I., Zhu, X., Ryan, M., Macalinao, D.G., Sousa, G.L., Caddle, L.B.,
Macnicoll, K.H., Barbay, J.M., Porciatti, V., Anderson, M.G., Smith, R.S., Clark, A.F.,
Libby, R.T., John, S.W., 2012. Radiation treatment inhibits monocyte entry into the
optic nerve head and prevents neuronal damage in a mouse model of glaucoma.
J. Clin. Invest. 122, 12461261.
Hu, H., Lu, W., Zhang, M., Zhang, X., Argall, A.J., Patel, S., Lee, G.E., Kim, Y.C.,
Jacobson, K.A., Laties, A.M., Mitchell, C.H., 2010. Stimulation of the P2X7 receptor
kills rat retinal ganglion cells in vivo. Exp. Eye Res. 91, 425432.
Huang, C., Han, X., Li, X., Lam, E., Peng, W., Lou, N., Torres, A., Yang, M., Garre, J.M.,
Tian, G.-F., Bennett, M.V.L., Nedergaard, M., Takano, T., 2012. Critical role of
connexin 43 in secondary expansion of traumatic spinal cord injury. J. Neurosci. 32,
33333338.
Iadecola, C., 2017. The neurovascular unit coming of age: a journey through
neurovascular coupling in health and disease. Neuron 96, 1742.
Ido, Y., Chang, K., Woolsey, T.A., Williamson, J.R., 2001. NADH: sensor of blood ow
need in brain, muscle, and other tissues. FASEB J. 15, 14191421.
Iglesias, R., Dahl, G., Qiu, F., Spray, D.C., Scemes, E., 2009. Pannexin 1: the molecular
substrate of astrocyte "hemichannels". J. Neurosci. 29, 70927097.
Ingerman-Wojenski, C., Silver, M.J., Smith, J.B., Macarak, E., 1981. Bovine endothelial
cells in culture produce thromboxane as well as prostacyclin. J. Clin. Invest. 67,
12921296.
Inman, D.M., Harun-or-Rashid, M., 2017. Metabolic vulnerability in the
neurodegenerative disease glaucoma. Front. Neurosci. 11, 146.
Inoue, A., Yanagisawa, M., Kimura, S., Kasuya, Y., Miyauchi, T., Goto, K., Masaki, T.,
1989. The human endothelin family: three structurally and pharmacologically
distinct isopeptides predicted by three separate genes. Proc. Natl. Acad. Sci. U. S. A.
86, 28632867.
Ishii, K., Kaneda, M., Li, H., Rockland, K.S., Hashikawa, T., 2003. Neuron-specic
distribution of P2X7 purinergic receptors in the monkey retina. J. Comp. Neurol.
459, 267277.
Ito, Y.A., Di Polo, A., 2017. Mitochondrial dynamics, transport, and quality control: a
bottleneck for retinal ganglion cell viability in optic neuropathies. Mitochondrion
36, 186192.
Ivanova, E., Corona, C., Eleftheriou, C.G., Bianchimano, P., Sagdullaev, B.T., 2021.
Retina-specic targeting of pericytes reveals structural diversity and enables control
of capillary blood ow. J. Comp. Neurol. 529, 11211134.
Iwase, C., Iwase, T., Tomita, R., Akahori, T., Yamamoto, K., Ra, E., Terasaki, H., 2021.
Changes in pulse waveforms in response to intraocular pressure elevation
determined by laser speckle owgraphy in healthy subjects. BMC Ophthalmol. 21,
303.
Janssen-Bienhold, U., Dermietzel, R., Weiler, R., 1998. Distribution of connexin43
immunoreactivity in the retinas of different vertebrates. J. Comp. Neurol. 396,
310321.
Jasty, U., Harris, A., Siesky, B., Rowe, L.W., Verticchio Vercellin, A.C., Mathew, S.,
Pasquale, L.R., 2020. Optic disc haemorrhage and primary open-angle glaucoma: a
clinical review. Br. J. Ophthalmol. 104, 14881491.
Jean-Louis, S., Lovasik, J.V., Kergoat, H., 2005. Systemic hyperoxia and retinal
vasomotor responses. Invest. Ophthalmol. Vis. Sci. 46, 17141720.
Jia, Y., Morrison, J.C., Tokayer, J., Tan, O., Lombardi, L., Baumann, B., Lu, C.D.,
Choi, W., Fujimoto, J.G., Huang, D., 2012. Quantitative OCT angiography of optic
nerve head blood ow. Biomed. Opt. Express 3, 31273137.
Jia, Y., Wei, E., Wang, X., Zhang, X., Morrison, J.C., Parikh, M., Lombardi, L.H.,
Gattey, D.M., Armour, R.L., Edmunds, B., Kraus, M.F., Fujimoto, J.G., Huang, D.,
2014. Optical coherence tomography angiography of optic disc perfusion in
glaucoma. Ophthalmology 121, 13221332.
Joly, S., Pernet, V., Chemtob, S., Di Polo, A., Lachapelle, P., 2007. Neuroprotection in the
juvenile rat model of light-induced retinopathy: evidence suggesting a role for FGF-2
and CNTF. Invest. Ophthalmol. Vis. Sci. 48, 23112320.
Jonas, J.B., Wang, N., Yang, D., Ritch, R., Panda-Jonas, S., 2015. Facts and myths of
cerebrospinal uid pressure for the physiology of the eye. Prog. Retin. Eye Res. 46,
6783.
Kawamura, H., Oku, H., Li, Q., Sakagami, K., Puro, D.G., 2002. Endothelin-induced
changes in the physiology of retinal pericytes. Invest. Ophthalmol. Vis. Sci. 43,
882888.
Kawarai, M., Koss, M.C., 1998. Sympathetic vasoconstriction in the rat anterior choroid
is mediated by alpha1-adrenoceptors. Eur. J. Pharmacol. 363, 3540.
Kerr, N.M., Johnson, C.S., De Souza, C.F., Chee, K.S., Good, W.R., Green, C.R., Danesh-
Meyer, H.V., 2010. Immunolocalization of gap junction protein connexin43 (GJA1)
in the human retina and optic nerve. Invest. Ophthalmol. Vis. Sci. 51, 40284034.
Kerr, N.M., Johnson, C.S., Green, C.R., Danesh-Meyer, H.V., 2011. Gap junction protein
connexin43 (GJA1) in the human glaucomatous optic nerve head and retina. J. Clin.
Neurosci. 18, 102108.
Kerr, N.M., Johnson, C.S., Zhang, J., Eady, E.K., Green, C.R., Danesh-Meyer, H.V., 2012.
High pressure-induced retinal ischaemia reperfusion causes upregulation of gap
junction protein connexin43 prior to retinal ganglion cell loss. Exp. Neurol. 234,
144152.
Kim, K.E., Kim, D.M., Flammer, J., Kim, K.N., 2015. Central retinal venous pressure in
eyes of normal-tension glaucoma patients with optic disc hemorrhage. PLoS One 10,
e0127920.
Kim, K.J., Ramiro Diaz, J., Iddings, J.A., Filosa, J.A., 2016. Vasculo-neuronal coupling:
retrograde vascular communication to brain nmooreeurons. J. Neurosci. 36,
1262412639.
Kimball, E.C., Jefferys, J.L., Pease, M.E., Oglesby, E.N., Nguyen, C., Schaub, J., Pitha, I.,
Quigley, H.A., 2018. The effects of age on mitochondria, axonal transport, and
axonal degeneration after chronic IOP elevation using a murine ocular explant
model. Exp. Eye Res. 172, 7885.
Kisler, K., Nelson, A.R., Rege, S.V., Ramanathan, A., Wang, Y., Ahuja, A., Lazic, D.,
Tsai, P.S., Zhao, Z., Zhou, Y., Boas, D.A., Sakadˇ
zi´
c, S., Zlokovic, B.V., 2017. Pericyte
degeneration leads to neurovascular uncoupling and limits oxygen supply to brain.
Nat. Neurosci. 20, 406416.
Kiss, B., Polska, E., Dorner, G., Polak, K., Findl, O., Mayrl, G.F., Eichler, H.G., Wolzt, M.,
Schmetterer, L., 2002. Retinal blood ow during hyperoxia in humans revisited:
concerted results using different measurement techniques. Microvasc. Res. 64,
7585.
Kiyota, N., Shiga, Y., Omodaka, K., Pak, K., Nakazawa, T., 2021. Time-course changes in
optic nerve head blood ow and retinal nerve ber layer thickness in eyes with open-
angle glaucoma. Ophthalmology 128, 663671.
Kiyota, N., Shiga, Y., Suzuki, S., Sato, M., Takada, N., Maekawa, S., Omodaka, K.,
Maruyama, K., Kunikata, H., Nakazawa, T., 2017. The effect of systemic hyperoxia
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
20
on optic nerve head blood ow in primary open-angle glaucoma patients. Invest.
Ophthalmol. Vis. Sci. 58, 31813188.
Kiyota, N., Shiga, Y., Yasuda, M., Aizawa, N., Omodaka, K., Tsuda, S., Pak, K.,
Kunikata, H., Nakazawa, T., 2020. The optic nerve head vasoreactive response to
systemic hyperoxia and visual eld defect progression in open-angle glaucoma, a
pilot study. Acta Ophthalmol. 98, e747e753.
Klose, E., Kuhrt, H., Kohen, L., Wiedemann, P., Bringmann, A., Hollborn, M., 2021.
Hypoxic and osmotic expression of Kir2.1 potassium channels in retinal pigment
epithelial cells: contribution to vascular endothelial growth factor expression. Exp.
Eye Res. 211, 108741.
Ko, K.R., Ngai, A.C., Winn, H.R., 1990. Role of adenosine in regulation of regional
cerebral blood ow in sensory cortex. Am. J. Physiol. 259, H1703H1708.
Koide, M., Moshkforoush, A., Tsoukias, N.M., Hill-Eubanks, D.C., Wellman, G.C.,
Nelson, M.T., Dabertrand, F., 2018. The yin and yang of K(V) channels in cerebral
small vessel pathologies. Microcirculation 25.
Kondo, M., Wang, L., Bill, A., 1997. The role of nitric oxide in hyperaemic response to
icker in the retina and optic nerve in cats. Acta Ophthalmol. Scand. 75, 232235.
Korn, C., Augustin, Hellmut g., 2015. Mechanisms of vessel pruning and regression. Dev.
Cell 34, 517.
Korneld, T.E., Newman, E.A., 2014. Regulation of blood ow in the retinal trilaminar
vascular network. J. Neurosci. 34, 1150411513.
Koto, T., Takubo, K., Ishida, S., Shinoda, H., Inoue, M., Tsubota, K., Okada, Y., Ikeda, E.,
2007. Hypoxia disrupts the barrier function of neural blood vessels through changes
in the expression of claudin-5 in endothelial cells. Am. J. Pathol. 170, 13891397.
Koukoula, S.C., Katsanos, A., Tentes, I.K., Labiris, G., Kozobolis, V.P., 2018. Retrobulbar
hemodynamics and aqueous humor levels of endothelin-1 in exfoliation syndrome
and exfoliation glaucoma. Clin. Ophthalmol. 12, 11991204.
Kovacs-Oller, T., Ivanova, E., Bianchimano, P., Sagdullaev, B.T., 2020. The pericyte
connectome: spatial precision of neurovascular coupling is driven by selective
connectivity maps of pericytes and endothelial cells and is disrupted in diabetes. Cell
Discovery 6, 39.
Kriˇ
zaj, D., Cordeiro, S., Strauß, O., 2023. Retinal TRP channels: cell-type-specic
regulators of retinal homeostasis and multimodal integration. Prog. Retin. Eye Res.
92, 101114.
Krüger-Genge, A., Blocki, A., Franke, R.P., Jung, F., 2019. Vascular endothelial cell
biology: an update. Int. J. Mol. Sci. 20.
Kugler, E.C., Greenwood, J., Macdonald, R.B., 2021. The "neuro-glial-vascular" unit: the
role of glia in neurovascular unit formation and dysfunction. Front. Cell Dev. Biol. 9,
732820.
Kur, J., Newman, E.A., Chan-Ling, T., 2012. Cellular and physiological mechanisms
underlying blood ow regulation in the retina and choroid in health and disease.
Prog. Retin. Eye Res. 31, 377406.
Kureli, G., Yilmaz-Ozcan, S., Erdener, S.E., Donmez-Demir, B., Yemisci, M., Karatas, H.,
Dalkara, T., 2020. F-actin polymerization contributes to pericyte contractility in
retinal capillaries. Exp. Neurol. 332, 113392.
Kustova, Y., Grinberg, A., Basile, A.S., 1999. Increased blood-brain barrier permeability
in LP-BM5 infected mice is mediated by neuroexcitatory mechanisms. Brain Res.
839, 153163.
Lander, H.M., Milbank, A.J., Tauras, J.M., Hajjar, D.P., Hempstead, B.L., Schwartz, G.D.,
Kraemer, R.T., Mirza, U.A., Chait, B.T., Burk, S.C., Quilliam, L.A., 1996. Redox
regulation of cell signalling. Nature 381, 380381.
Laties, A.M., 1967. Central retinal artery innervation. Absence of adrenergic innervation
to the intraocular branches. Arch. Ophthalmol. 77, 405409.
Lau, J., Dang, M., Hockmann, K., Ball, A.K., 2006. Effects of acute delivery of endothelin-
1 on retinal ganglion cell loss in the rat. Exp. Eye Res. 82, 132145.
Leal-Campanario, R., Alarcon-Martinez, L., Rieiro, H., Martinez-Conde, S., Alarcon-
Martinez, T., Zhao, X., Lamee, J., Popp, P.J., Calhoun, M.E., Arribas, J.I., Schlegel, A.
A., Stasi, L.L., Rho, J.M., Inge, L., Otero-Millan, J., Treiman, D.M., Macknik, S.L.,
2017. Abnormal capillary vasodynamics contribute to ictal neurodegeneration in
epilepsy. Sci. Rep. 7, 43276.
Lebrun-Julien, F., Bertrand, M., De Backer, O., Stellwagen, D., Morales, C.R., Di Polo, A.,
Barker, P.A., 2010. ProNGF induces TNF
α
-dependent death of retinal ganglion cells
through a p75NTR non-cell-autonomous signaling pathway. Proc. Natl. Acad. Sci. U.
S. A. 107, 38173822.
Lebrun-Julien, F., Duplan, L., Pernet, V., Osswald, I.K., Sapieha, P., Bourgeois, P.,
Dickson, K., Bowie, D., Barker, P.A., Di Polo, A., 2009a. Excitotoxic death of retinal
neurons in vivo occurs via a non-cell-autonomous mechanism. J. Neurosci. 29,
55365545.
Lebrun-Julien, F., Morquette, B., Douillette, A., Saragovi, H.U., Di Polo, A., 2009b.
Inhibition of p75NTR in glia potentiates TrkA-mediated survival of injured retinal
ganglion cells. Mol. Cell. Neurosci. 40, 410420.
Lecoq, J., Parpaleix, A., Roussakis, E., Ducros, M., Goulam Houssen, Y., Vinogradov, S.A.,
Charpak, S., 2011. Simultaneous two-photon imaging of oxygen and blood ow in
deep cerebral vessels. Nat. Med. 17, 893898.
Lecrux, C., Hamel, E., 2016. Neuronal networks and mediators of cortical neurovascular
coupling responses in normal and altered brain states. Philos. Trans. R. Soc. Lond. B
Biol. Sci. 371.
Lee, E.J., Han, J.C., Kee, C., 2017a. A novel hypothesis for the pathogenesis of
glaucomatous disc hemorrhage. Prog. Retin. Eye Res. 60, 2043.
Lee, E.J., Lee, K.M., Lee, S.H., Kim, T.W., 2017b. Parapapillary choroidal
microvasculature dropout in glaucoma: a comparison between optical coherence
tomography angiography and indocyanine green angiography. Ophthalmology 124,
12091217.
Lee, E.J., Lee, S.H., Kim, J.A., Kim, T.W., 2017c. Parapapillary deep-Layer
microvasculature dropout in glaucoma: topographic association with glaucomatous
damage. Invest. Ophthalmol. Vis. Sci. 58, 30043010.
Lee, H.S., Jun, J.H., Jung, E.H., Koo, B.A., Kim, Y.S., 2014. Epigalloccatechin-3-gallate
inhibits ocular neovascularization and vascular permeability in human retinal
pigment epithelial and human retinal microvascular endothelial cells via suppression
of MMP-9 and VEGF activation. Molecules 19, 1215012172.
Lee, P.Y., Hua, Y., Brazile, B.L., Yang, B., Wang, L., Sigal, I.A., 2022. A workow for
three-dimensional reconstruction and quantication of the monkey optic nerve head
vascular network. J. Biomech. Eng. 144.
Leske, M.C., 2009. Ocular perfusion pressure and glaucoma: clinical trial and
epidemiologic ndings. Curr. Opin. Ophthalmol. 20, 7378.
Leske, M.C., Heijl, A., Hyman, L., Bengtsson, B., Dong, L., Yang, Z., Group, E., 2007.
Predictors of long-term progression in the early manifest glaucoma trial.
Ophthalmology 114, 19651972.
Leske, M.C., Wu, S.Y., Hennis, A., Honkanen, R., Nemesure, B., Group, B.E.S., 2008. Risk
factors for incident open-angle glaucoma: the Barbados Eye Studies. Ophthalmology
115, 8593.
Leybaert, L., Sanderson, M.J., 2012. Intercellular Ca(2+) waves: mechanisms and
function. Physiol. Rev. 92, 13591392.
Li, A., Zhang, X., Zheng, D., Ge, J., Laties, A.M., Mitchell, C.H., 2011. Sustained elevation
of extracellular ATP in aqueous humor from humans with primary chronic angle-
closure glaucoma. Exp. Eye Res. 93, 528533.
Li, F., Jiang, D., Samuel, M.A., 2019. Microglia in the developing retina. Neural Dev. 14,
12.
Li, Q., Puro, D.G., 2001. Adenosine activates ATP-sensitive K(+) currents in pericytes of
rat retinal microvessels: role of A1 and A2a receptors. Brain Res. 907, 9399.
Li, T., Li, D., Wei, Q., Shi, M., Xiang, J., Gao, R., Chen, C., Xu, Z.X., 2023. Dissecting the
neurovascular unit in physiology and Alzheimers disease: functions, imaging tools
and genetic mouse models. Neurobiol. Dis. 181, 106114.
Liang, Y., Fortune, B., Cull, G., Ciof, G.A., Wang, L., 2010. Quantication of dynamic
blood ow autoregulation in optic nerve head of rhesus monkeys. Exp. Eye Res. 90,
203209.
Libby, R., Howell, G., Pang, I.-H., Savinova, O., Mehalow, A., Barter, J., Smith, R.,
Clark, A., John, S., 2007. Inducible nitric oxide synthase, Nos2, does not mediate
optic neuropathy and retinopathy in the DBA/2J glaucoma model. BMC Neurosci. 8,
108.
Lichter, P.R., Henderson, J.W., 1978. Optic nerve infarction. Am. J. Ophthalmol. 85,
302310.
Lindahl, P., Johansson, B.R., Leveen, P., Betsholtz, C., 1997. Pericyte loss and
microaneurysm formation in PDGF-B-decient mice. Science 277, 242245.
Lindauer, U., Megow, D., Matsuda, H., Dirnagl, U., 1999. Nitric oxide: a modulator, but
not a mediator, of neurovascular coupling in rat somatosensory cortex. Am. J.
Physiol. 277, H799H811.
Lindblom, P., Gerhardt, H., Liebner, S., Abramsson, A., Enge, M., Hellstr¨
om, M.,
B¨
ackstr¨
om, G., Fredriksson, S., Landegren, U., Nystr¨
om, H.C., Bergstr¨
om, G.,
Dejana, E., ¨
Ostman, A., Lindahl, P., Betsholtz, C., 2003. Endothelial PDGF-B
retention is required for proper investment of pericytes in the microvessel wall.
Genes Dev. 17, 18351840.
Liu, H.-T., Toychiev, A.H., Takahashi, N., Sabirov, R.Z., Okada, Y., 2008. Maxi-anion
channel as a candidate pathway for osmosensitive ATP release from mouse
astrocytes in primary culture. Cell Res. 18, 558565.
Liu, L., Edmunds, B., Takusagawa, H.L., Tehrani, S., Lombardi, L.H., Morrison, J.C.,
Jia, Y., Huang, D., 2019. Projection-resolved optical coherence tomography
angiography of the peripapillary retina in glaucoma. Am. J. Ophthalmol. 207,
99109.
Liu, L., Jia, Y., Takusagawa, H.L., Pechauer, A.D., Edmunds, B., Lombardi, L., Davis, E.,
Morrison, J.C., Huang, D., 2015. Optical coherence tomography angiography of the
peripapillary retina in glaucoma. JAMA Ophthalmol 133, 10451052.
Loebl, M., Schwartz, B., 1977. Fluorescein angiographic defects of the optic disc in ocular
hypertension. Arch. Ophthalmol. 95, 19801984.
Loh, Y.C., Tan, C.S., Chng, Y.S., Yeap, Z.Q., Ng, C.H., Yam, M.F., 2018. Overview of the
microenvironment of vasculature in vascular tone regulation. Int. J. Mol. Sci. 19.
Lohman, A.W., Isakson, B.E., 2014. Differentiating connexin hemichannels and pannexin
channels in cellular ATP release. FEBS Lett. 588, 13791388.
Longden, T.A., Dabertrand, F., Hill-Eubanks, D.C., Hammack, S.E., Nelson, M.T., 2014.
Stress-induced glucocorticoid signaling remodels neurovascular coupling through
impairment of cerebrovascular inwardly rectifying K+channel function. Proc. Natl.
Acad. Sci. U. S. A. 111, 74627467.
Longden, T.A., Dabertrand, F., Koide, M., Gonzales, A.L., Tykocki, N.R., Brayden, J.E.,
Hill-Eubanks, D., Nelson, M.T., 2017. Capillary K(+)-sensing initiates retrograde
hyperpolarization to increase local cerebral blood ow. Nat. Neurosci. 20, 717726.
Longden, T.A., Hill-Eubanks, D.C., Nelson, M.T., 2016. Ion channel networks in the
control of cerebral blood ow. J. Cerebr. Blood Flow Metabol. 36, 492512.
Longden, T.A., Mughal, A., Hennig, G.W., Harraz, O.F., Shui, B., Lee, F.K., Lee, J.C.,
Reining, S., Kotlikoff, M.I., K¨
onig, G.M., Kostenis, E., Hill-Eubanks, D., Nelson, M.T.,
2021. Local IP3 receptor-mediated Ca2+signals compound to direct blood ow in
brain capillaries. Sci. Adv. 7, eabh0101.
Longden, T.A., Nelson, M.T., 2015. Vascular inward rectier K+channels as external K+
sensors in the control of cerebral blood ow. Microcirculation 22, 183196.
Longden, T.A., Zhao, G., Hariharan, A., Lederer, W.J., 2023. Pericytes and the control of
blood ow in brain and heart. Annu. Rev. Physiol. 85, 137164.
Lou, N., Takano, T., Pei, Y., Xavier, A.L., Goldman, S.A., Nedergaard, M., 2016.
Purinergic receptor P2RY12-dependent microglial closure of the injured blood-brain
barrier. Proc. Natl. Acad. Sci. U. S. A. 113, 10741079.
Lu, W., Hu, H., S´
evigny, J., Gabelt, B.T., Kaufman, P.L., Johnson, E.C., Morrison, J.C.,
Zode, G.S., Shefeld, V.C., Zhang, X., Laties, A.M., Mitchell, C.H., 2015. Rat, mouse,
and primate models of chronic glaucoma show sustained elevation of extracellular
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
21
ATP and altered purinergic signaling in the posterior eye. Invest. Ophthalmol. Vis.
Sci. 56, 30753083.
Luft, N., Wozniak, P.A., Aschinger, G.C., Fondi, K., Bata, A.M., Werkmeister, R.M.,
Schmidl, D., Witkowska, K.J., Bolz, M., Garh¨
ofer, G., Schmetterer, L., 2016. Ocular
blood ow measurements in healthy white subjects using laser speckle owgraphy.
PLoS One 11, e0168190.
Maccumber, M.W., Danna, S.A., 1994. Endothelin receptor-binding subtypes in the
human retina and choroid. Arch. Ophthalmol. 112, 12311235.
Mackenzie, P.J., Ciof, G.A., 2008. Vascular anatomy of the optic nerve head. Can. J.
Ophthalmol. 43, 308312.
M¨
ae, M.A., He, L., Nordling, S., Vazquez-Liebanas, E., Nahar, K., Jung, B., Li, X., Tan, B.
C., Chin Foo, J., Cazenave-Gassiot, A., Wenk, M.R., Zarb, Y., Lavina, B., Quaggin, S.
E., Jeansson, M., Gu, C., Silver, D.L., Vanlandewijck, M., Butcher, E.C., Keller, A.,
Betsholtz, C., 2021. Single-cell analysis of blood-brain barrier response to pericyte
loss. Circ. Res. 128, e46e62.
Maekawa, S., Shiga, Y., Kawasaki, R., Nakazawa, T., 2014. Usefulness of novel laser
speckle owgraphy-derived variables of the large vessel area in the optic nerve head
in normal tension glaucoma. Clin. Exp. Ophthalmol. 42, 887889.
Mandarino, L.J., Sundarraj, N., Finlayson, J., Hassell, H.R., 1993. Regulation of
bronectin and laminin synthesis by retinal capillary endothelial cells and pericytes
in vitro. Exp. Eye Res. 57, 609621.
Mansoori, T., Sivaswamy, J., Gamalapati, J.S., Balakrishna, N., 2017. Radial
peripapillary capillary density measurement using optical coherence tomography
angiography in early glaucoma. J. Glaucoma 26, 438443.
Mao, Y., Tonkin, R.S., Nguyen, T., Ocarroll, S.J., Nicholson, L.F.B., Green, C.R., Moalem-
Taylor, G., Gorrie, C.A., 2017. Systemic administration of connexin43 mimetic
peptide improves functional recovery after traumatic spinal cord injury in adult rats.
J. Neurotrauma 34, 707719.
Marola, O.J., Howell, G.R., Libby, R.T., 2022. Vascular derived endothelin receptor A
controls endothelin-induced retinal ganglion cell death. Cell Death Dis. 8, 207.
Marola, O.J., Syc-Mazurek, S.B., Howell, G.R., Libby, R.T., 2020. Endothelin 1-induced
retinal ganglion cell death is largely mediated by JUN activation. Cell Death Dis. 11,
811.
Mat Nor, M.N., Rupenthal, I.D., Green, C.R., Acosta, M.L., 2020. Connexin hemichannel
block using orally delivered tonabersat improves outcomes in animal models of
retinal disease. Neurotherapeutics 17, 371387.
Maynard, E.A., Schultz, R.L., Pease, D.C., 1957. Electron microscopy of the vascular bed
of rat cerebral cortex. Am. J. Anat. 100, 409433.
Mendes-Jorge, L., Llombart, C., Ramos, D., L´
opez-Luppo, M., Valença, A., Nacher, V.,
Navarro, M., Carretero, A., M´
endez-Ferrer, S., Rodriguez-Baeza, A., Ruberte, J.,
2012. Intercapillary bridging cells: immunocytochemical characteristics of cells that
connect blood vessels in the retina. Exp. Eye Res. 98, 7987.
Metea, M.R., Newman, E.A., 2006a. Calcium signaling in specialized glial cells. Glia 54,
650655.
Metea, M.R., Newman, E.A., 2006b. Glial cells dilate and constrict blood vessels: a
mechanism of neurovascular coupling. J. Neurosci. 26, 28622870.
Mi, X.S., Zhang, X., Feng, Q., Lo, A.C., Chung, S.K., So, K.F., 2012. Progressive retinal
degeneration in transgenic mice with overexpression of endothelin-1 in vascular
endothelial cells. Invest. Ophthalmol. Vis. Sci. 53, 48424851.
Mills, S.A., Jobling, A.I., Dixon, M.A., Bui, B.V., Vessey, K.A., Phipps, J.A., Greferath, U.,
Venables, G., Wong, V.H.Y., Wong, C.H.Y., He, Z., Hui, F., Young, J.C., Tonc, J.,
Ivanova, E., Sagdullaev, B.T., Fletcher, E.L., 2021. Fractalkine-induced microglial
vasoregulation occurs within the retina and is altered early in diabetic retinopathy.
Proc. Natl. Acad. Sci. U. S. A. 118.
Minckler, D.S., Bunt, A.H., 1977. Axoplasmic transport in ocular hypotony and
papilledema in the monkey. Arch. Ophthalmol. 95, 14301436.
Minton, A.Z., Phatak, N.R., Stankowska, D.L., He, S., Ma, H.Y., Mueller, B.H., Jiang, M.,
Luedtke, R., Yang, S., Brownlee, C., Krishnamoorthy, R.R., 2012. Endothelin B
receptors contribute to retinal ganglion cell loss in a rat model of glaucoma. PLoS
One 7, e43199.
Misfeldt, M.W., Pedersen, S.M., Bek, T., 2013. Perivascular cells with pericyte
characteristics are involved in ATP- and PGE(2)-induced relaxation of porcine retinal
arterioles in vitro. Invest. Ophthalmol. Vis. Sci. 54, 32583264.
Mishra, A., Hamid, A., Newman, E.A., 2011. Oxygen modulation of neurovascular
coupling in the retina. Proc. Natl. Acad. Sci. U. S. A. 108, 1782717831.
Mishra, A., Newman, E.A., 2010. Inhibition of inducible nitric oxide synthase reverses
the loss of functional hyperemia in diabetic retinopathy. Glia 58, 19962004.
Mittal, R., Karhu, E., Wang, J.S., Delgado, S., Zukerman, R., Mittal, J., Jhaveri, V.M.,
2019. Cell communication by tunneling nanotubes: implications in disease and
therapeutic applications. J. Cell. Physiol. 234, 11301146.
Moghimi, S., Zangwill, L.M., Penteado, R.C., Hasenstab, K., Ghahari, E., Hou, H.,
Christopher, M., Yarmohammadi, A., Manalastas, P.I.C., Shoji, T., Bowd, C.,
Weinreb, R.N., 2018. Macular and optic nerve head vessel density and progressive
retinal nerve ber layer loss in glaucoma. Ophthalmology 125, 17201728.
Montagne, A., Zhao, Z., Zlokovic, B.V., 2017. Alzheimers disease: a matter of blood-
brain barrier dysfunction? J. Exp. Med. 214, 31513169.
Moore, C.I., Cao, R., 2008. The hemo-neural hypothesis: on the role of blood ow in
information processing. J. Neurophysiol. 99, 20352047.
Mori, A., Saito, M., Sakamoto, K., Narita, M., Nakahara, T., Ishii, K., 2007. Stimulation of
prostanoid IP and EP(2) receptors dilates retinal arterioles and increases retinal and
choroidal blood ow in rats. Eur. J. Pharmacol. 570, 135141.
Mori, A., Yano, E., Sakamoto, K., Ishii, K., Nakahara, T., 2021. Role of
epoxyeicosatrienoic acids in acetylcholine-induced dilation of rat retinal arterioles in
vivo. Biol. Pharm. Bull. 44, 8287.
Morquette, J.B., Di Polo, A., 2008. Dendritic and synaptic protection: is it enough to save
the retinal ganglion cell body and axon? J. Neuro Ophthalmol. 28, 144154.
Moshkforoush, A., Ashenagar, B., Harraz, O.F., Dabertrand, F., Longden, T.A., Nelson, M.
T., Tsoukias, N.M., 2020. The capillary Kir channel as sensor and amplier of
neuronal signals: modeling insights on K+-mediated neurovascular communication.
Proc. Natl. Acad. Sci. U. S. A. 117, 1662616637.
Mosso, A., 1880. Sulla circolazione del sangue nel cervello delluomo. R. Accad. Lincei 5,
237358.
Moyaert, P., Padrela, B.E., Morgan, C.A., Petr, J., Versijpt, J., Barkhof, F., Jurkiewicz, M.
T., Shao, X., Oyeniran, O., Manson, T., Wang, D.J.J., Günther, M., Achten, E.,
Mutsaerts, H., Anazodo, U.C., 2023. Imaging blood-brain barrier dysfunction: a
state-of-the-art review from a clinical perspective. Front. Aging Neurosci. 15,
1132077.
Mugisho, O.O., Green, C.R., Kho, D.T., Zhang, J., Graham, E.S., Acosta, M.L.,
Rupenthal, I.D., 2018. The inammasome pathway is amplied and perpetuated in
an autocrine manner through connexin43 hemichannel mediated ATP release.
Biochim. Biophys. Acta 1862, 385393.
Mulligan, S.J., Macvicar, B.A., 2004. Calcium transients in astrocyte endfeet cause
cerebrovascular constrictions. Nature 431, 195199.
Murakami, M., Sugita, A., Shimada, T., Nakamura, K., 1979. Surface view of pericytes on
the retinal capillary in rabbits revealed by scanning electron microscopy. Arch.
Histol. Jpn. 42, 297303.
Muthusamy, A., Lin, C.M., Shanmugam, S., Lindner, H.M., Abcouwer, S.F., Antonetti, D.
A., 2014. Ischemia-reperfusion injury induces occludin phosphorylation/
ubiquitination and retinal vascular permeability in a VEGFR-2-dependent manner.
J. Cerebr. Blood Flow Metabol. 34, 522531.
Nagel, E., Vilser, W., Lanzl, I.M., Lanzi, I.M., 2001. Retinal vessel reaction to short-term
IOP elevation in ocular hypertensive and glaucoma patients. Eur. J. Ophthalmol. 11,
338344.
Nampoothiri, S., Nogueiras, R., Schwaninger, M., Prevot, V., 2022. Glial cells as
integrators of peripheral and central signals in the regulation of energy homeostasis.
Nat. Metab. 4, 813825.
Nanba, K., Schwartz, B., 1988. Nerve ber layer and optic disc uorescein defects in
glaucoma and ocular hypertension. Ophthalmology 95, 12271233.
Nehls, V., Drenckhahn, D., 1991. Heterogeneity of microvascular pericytes for smooth
muscle type alpha-actin. J. Cell Biol. 113, 147154.
Nelson, A.R., Sagare, M.A., Wang, Y., Kisler, K., Zhao, Z., Zlokovic, B.V., 2020.
Channelrhodopsin excitation contracts brain pericytes and reduces blood ow in the
aging mouse brain in vivo. Front. Aging Neurosci. 12, 108.
Neufeld, A.H., Hernandez, M.R., Gonzalez, M., 1997. Nitric oxide synthase in the human
glaucomatous optic nerve head. Arch. Ophthalmol. 115, 497503.
Neufeld, A.H., Sawada, A., Becker, B., 1999. Inhibition of nitric-oxide synthase 2 by
aminoguanidine provides neuroprotection of retinal ganglion cells in a rat model of
chronic glaucoma. Proc. Natl. Acad. Sci. U.S.A. 96, 99449948.
Newman, A., Andrew, N., Casson, R., 2018. Review of the association between retinal
microvascular characteristics and eye disease. Clin. Exp. Ophthalmol. 46, 531552.
Newman, E., Reichenbach, A., 1996. The Muller cell: a functional element of the retina.
Trends Neurosci. 19, 307312.
Newman, E.A., 2013. Functional hyperemia and mechanisms of neurovascular coupling
in the retinal vasculature. J. Cerebr. Blood Flow Metabol. 33, 16851695.
Newman, E.A., 2015. Glial cell regulation of neuronal activity and blood ow in the
retina by release of gliotransmitters. Philos. Trans. R. Soc. Lond. B Biol. Sci. 370,
20140195.
Nian, S., Lo, A.C.Y., Mi, Y., Ren, K., Yang, D., 2021. Neurovascular unit in diabetic
retinopathy: pathophysiological roles and potential therapeutical targets. Eye Vis. 8,
15.
Nickells, R.W., Howell, G.R., Soto, I., John, S.W.M., 2012. Under pressure: cellular and
molecular responses during glaucoma, a common neurodegeneration with
axonopathy. Annu. Rev. Neurosci. 35, 153179.
Nikolakopoulou, A.M., Montagne, A., Kisler, K., Dai, Z., Wang, Y., Huuskonen, M.T.,
Sagare, A.P., Lazic, D., Sweeney, M.D., Kong, P., Wang, M., Owens, N.C., Lawson, E.
J., Xie, X., Zhao, Z., Zlokovic, B.V., 2019. Pericyte loss leads to circulatory failure
and pleiotrophin depletion causing neuron loss. Nat. Neurosci. 22, 10891098.
Nilsson, S.F., 1996. Nitric oxide as a mediator of parasympathetic vasodilation in ocular
and extraocular tissues in the rabbit. Invest. Ophthalmol. Vis. Sci. 37, 21102119.
Nippert, A.R., Newman, E.A., 2020. Regulation of blood ow in diabetic retinopathy. Vis.
Neurosci. 37, E004.
Nitta, T., Hata, M., Gotoh, S., Seo, Y., Sasaki, H., Hashimoto, N., Furuse, M., Tsukita, S.,
2003. Size-selective loosening of the blood-brain barrier in claudin-5-decient mice.
J. Cell Biol. 161, 653660.
Noack, F., Christ, M., May, S.A., Steinmeier, R., Morgenstern, U., 2007. Assessment of
dynamic changes in cerebral autoregulation. Biomed. Tech. 52, 3136.
Nortley, R., Korte, N., Izquierdo, P., Hirunpattarasilp, C., Mishra, A., Jaunmuktane, Z.,
Kyrargyri, V., Pfeiffer, T., Khennouf, L., Madry, C., Gong, H., Richard-Loendt, A.,
Huang, W., Saito, T., Saido, T.C., Brandner, S., Sethi, H., Attwell, D., 2019. Amyloid
β oligomers constrict human capillaries in Alzheimers disease via signaling to
pericytes. Science 365, eaav9518.
Ocarroll, S., Alkadhi, M., Nicholson, L.F., Green, C., 2008. Connexin 43 mimetic
peptides reduce swelling, astrogliosis, and neuronal cell death after spinal cord
injury. Cell Commun. Adhes. 15, 2742.
Ocarroll, S.J., Becker, D.L., Davidson, J.O., Gunn, A.J., Nicholson, L.F.B., Green, C.R.,
2013. The use of connexin-based therapeutic approaches to target inammatory
diseases. Methods Mol. Biol. 1037, 519546.
Ofarrell, F.M., Mastitskaya, S., Hammond-Haley, M., Freitas, F., Wah, W.R., Attwell, D.,
2017. Capillary pericytes mediate coronary no-reow after myocardial ischaemia.
Elife 6, e29280.
Oleary, F., Campbell, M., 2023. The bloodretina barrier in health and disease. FEBS J.
290, 878891.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
22
Ogura, S., Kurata, K., Hattori, Y., Takase, H., Ishiguro-Oonuma, T., Hwang, Y., Ahn, S.,
Park, I., Ikeda, W., Kusuhara, S., Fukushima, Y., Nara, H., Sakai, H., Fujiwara, T.,
Matsushita, J., Ema, M., Hirashima, M., Minami, T., Shibuya, M., Takakura, N.,
Kim, P., Miyata, T., Ogura, Y., Uemura, A., 2017. Sustained inammation after
pericyte depletion induces irreversible blood-retina barrier breakdown. JCI Insight
2, e90905.
Orgül, S., Ciof, G., Bacon, D., Van Buskirk, E., 1996. An endothelin-1-induced model of
chronic optic nerve ischemia in rhesus monkeys. J. Glaucoma 5, 135138.
Orgül, S., Gugleta, K., Flammer, J., 1999. Physiology of perfusion as it relates to the optic
nerve head. Surv. Ophthalmol. 43 (Suppl. 1), S17S26.
Ornelas, S., Berthiaume, A.A., Bonney, S.K., Coelho-Santos, V., Underly, R.G., Kremer, A.,
Gu´
erin, C.J., Lippens, S., Shih, A.Y., 2021. Three-dimensional ultrastructure of the
brain pericyte-endothelial interface. J. Cerebr. Blood Flow Metabol. 41, 21852200.
Ortin-Martinez, A., Yan, N.E., Tsai, E.L.S., Comanita, L., Gurdita, A., Tachibana, N.,
Liu, Z.C., Lu, S., Dolati, P., Pokrajac, N.T., El-Sehemy, A., Nickerson, P.E.B.,
Schuurmans, C., Bremner, R., Wallace, V.A., 2021. Photoreceptor nanotubes mediate
the in vivo exchange of intracellular material. EMBO J. 40, e107264.
Osborne, N.N., Nú˜
nez-´
Alvarez, C., Joglar, B., Del Olmo-Aguado, S., 2016. Glaucoma:
focus on mitochondria in relation to pathogenesis and neuroprotection. Eur. J.
Pharmacol. 787, 127133.
Osswald, M., Jung, E., Sahm, F., Solecki, G., Venkataramani, V., Blaes, J., Weil, S.,
Horstmann, H., Wiestler, B., Syed, M., Huang, L., Ratliff, M., Karimian Jazi, K.,
Kurz, F.T., Schmenger, T., Lemke, D., G¨
ommel, M., Pauli, M., Liao, Y., H¨
aring, P.,
Pusch, S., Herl, V., Steinh¨
auser, C., Krunic, D., Jarahian, M., Miletic, H., Berghoff, A.
S., Griesbeck, O., Kalamakis, G., Garaschuk, O., Preusser, M., Weiss, S., Liu, H.,
Heiland, S., Platten, M., Huber, P.E., Kuner, T., Von Deimling, A., Wick, W.,
Winkler, F., 2015. Brain tumour cells interconnect to a functional and resistant
network. Nature 528, 93.
Ott, M.J., Olson, J.L., Ballermann, B.J., 1995. Chronic in vitro ow promotes
ultrastructural differentiation of endothelial cells. Endothelium 3, 2130.
Oudemans-Van Straaten, H.M., Fiaccadori, E., Baldwin, I., 2010. Anticoagulation for
renal replacement therapy: different methods to improve safety. Contrib. Nephrol.
165, 251262.
Outtz, H.H., Tattersall, I.W., Koer, N.M., Steinbach, N., Kitajewski, J., 2011. Notch1
controls macrophage recruitment and Notch signaling is activated at sites of
endothelial cell anastomosis during retinal angiogenesis in mice. Blood 118,
34363439.
Panerai, R.B., 1998. Assessment of cerebral pressure autoregulation in humans–a review
of measurement methods. Physiol. Meas. 19, 305338.
Pang, I.-H., Johnson, E.C., Jia, L., Cepurna, W.O., Shepard, A.R., Hellberg, M.R., Clark, A.
F., Morrison, J.C., 2005. Evaluation of inducible nitric oxide synthase in
glaucomatous optic neuropathy and pressure-induced optic nerve damage. Invest.
Ophthalmol. Vis. Sci. 46, 13131321.
Paolicelli, R.C., Sierra, A., Stevens, B., Tremblay, M.E., Aguzzi, A., Ajami, B., Amit, I.,
Audinat, E., Bechmann, I., Bennett, M., Bennett, F., Bessis, A., Biber, K., Bilbo, S.,
Blurton-Jones, M., Boddeke, E., Brites, D., Brˆ
one, B., Brown, G.C., Butovsky, O.,
Carson, M.J., Castellano, B., Colonna, M., Cowley, S.A., Cunningham, C.,
Davalos, D., De Jager, P.L., De Strooper, B., Denes, A., Eggen, B.J.L., Eyo, U.,
Galea, E., Garel, S., Ginhoux, F., Glass, C.K., Gokce, O., Gomez-Nicola, D.,
Gonz´
alez, B., Gordon, S., Graeber, M.B., Greenhalgh, A.D., Gressens, P., Greter, M.,
Gutmann, D.H., Haass, C., Heneka, M.T., Heppner, F.L., Hong, S., Hume, D.A.,
Jung, S., Kettenmann, H., Kipnis, J., Koyama, R., Lemke, G., Lynch, M.,
Majewska, A., Malcangio, M., Malm, T., Mancuso, R., Masuda, T., Matteoli, M.,
Mccoll, B.W., Miron, V.E., Molofsky, A.V., Monje, M., Mracsko, E., Nadjar, A.,
Neher, J.J., Neniskyte, U., Neumann, H., Noda, M., Peng, B., Peri, F., Perry, V.H.,
Popovich, P.G., Pridans, C., Priller, J., Prinz, M., Ragozzino, D., Ransohoff, R.M.,
Salter, M.W., Schaefer, A., Schafer, D.P., Schwartz, M., Simons, M., Smith, C.J.,
Streit, W.J., Tay, T.L., Tsai, L.H., Verkhratsky, A., Von Bernhardi, R., Wake, H.,
Wittamer, V., Wolf, S.A., Wu, L.J., Wyss-Coray, T., 2022. Microglia states and
nomenclature: a eld at its crossroads. Neuron 110, 34583483.
Park, D.Y., Lee, J., Kim, J., Kim, K., Hong, S., Han, S., Kubota, Y., Augustin, H.G.,
Ding, L., Kim, J.W., Kim, H., He, Y., Adams, R.H., Koh, G.Y., 2017. Plastic roles of
pericytes in the bloodretinal barrier. Nat. Commun. 8, 15296.
Park, H.Y.L., Jeong, H.J., Kim, Y.H., Park, C.K., 2015. Optic disc hemorrhage is related to
various hemodynamic ndings by disc angiography. PLoS One 10, e0120000.
Parpaleix, A., Goulam Houssen, Y., Charpak, S., 2013. Imaging local neuronal activity by
monitoring POtransients in capillaries. Nat. Med. 19, 241246.
Patel, C., Narayanan, S.P., Zhang, W., Xu, Z., Sukumari-Ramesh, S., Dhandapani, K.M.,
Caldwell, R.W., Caldwell, R.B., 2014. Activation of the endothelin system mediates
pathological angiogenesis during ischemic retinopathy. Am. J. Pathol. 184,
30403051.
Peppiatt, C.M., Howarth, C., Mobbs, P., Attwell, D., 2006. Bidirectional control of CNS
capillary diameter by pericytes. Nature 443, 700704.
Per´
alvarez-Marín, A., Do˜
nate-Macian, P., Gaudet, R., 2013. What do we know about the
transient receptor potential vanilloid 2 (TRPV2) ion channel? FEBS J. 280,
54715487.
Pernet, V., Bourgeois, P., Di Polo, A., 2007. A role for polyamines in retinal ganglion cell
excitotoxic death. J. Neurochem. 103, 14811490.
Pfeiffer, F., Sch¨
afer, J., Lyck, R., Makrides, V., Brunner, S., Schaeren-Wiemers, N.,
Deutsch, U., Engelhardt, B., 2011. Claudin-1 induced sealing of blood-brain barrier
tight junctions ameliorates chronic experimental autoimmune encephalomyelitis.
Acta Neuropathol. 122, 601614.
Pfeiffer, T., Li, Y., Attwell, D., 2021. Diverse mechanisms regulating brain energy supply
at the capillary level. Curr. Opin. Neurobiol. 69, 4150.
Pillunat, K.R., Ventzke, S., Spoerl, E., Furashova, O., Stodtmeister, R., Pillunat, L.E.,
2014. Central retinal venous pulsation pressure in different stages of primary open-
angle glaucoma. Br. J. Ophthalmol. 98, 13741378.
Pillunat, L.E., Anderson, D.R., Knighton, R.W., Joos, K.M., Feuer, W.J., 1997.
Autoregulation of human optic nerve head circulation in response to increased
intraocular pressure. Exp. Eye Res. 64, 737744.
Pitts, K.M., Margeta, M.A., 2023. Myeloid masquerade: microglial transcriptional
signatures in retinal development and disease. Front. Cell. Neurosci. 17, 1106547.
Plange, N., Bienert, M., Remky, A., Arend, K.O., 2012. Optic disc uorescein leakage and
intraocular pressure in primary open-angle glaucoma. Curr. Eye Res. 37, 508512.
Plange, N., Kaup, M., Doehmen, B., Remky, A., Arend, K.O., 2010. Fluorescein leakage of
the optic disc: time course in primary open-angle glaucoma. Ophthalmic Physiol.
Opt. 30, 315320.
Plange, N., Kaup, M., Huber, K., Remky, A., Arend, O., 2006. Fluorescein lling defects of
the optic nerve head in normal tension glaucoma, primary open-angle glaucoma,
ocular hypertension and healthy controls. Ophthalmic Physiol. Opt. 26, 2632.
Polak, K., Schmetterer, L., Riva, C.E., 2002. Inuence of icker frequency on icker-
induced changes of retinal vessel diameter. Invest. Ophthalmol. Vis. Sci. 43,
27212726.
Prada, D., Harris, A., Guidoboni, G., Siesky, B., Huang, A.M., Arciero, J., 2016.
Autoregulation and neurovascular coupling in the optic nerve head. Surv.
Ophthalmol. 61, 164186.
Prasanna, G., Krishnamoorthy, R., Yorio, T., 2011. Endothelin, astrocytes and glaucoma.
Exp. Eye Res. 93, 170177.
Prinz, M., Masuda, T., Wheeler, M.A., Quintana, F.J., 2021. Microglia and central
nervous system-associated macrophages-from origin to disease modulation. Annu.
Rev. Immunol. 39, 251277.
Puro, D.G., 2007. Physiology and pathobiology of the pericyte-containing retinal
microvasculature: new developments. Microcirculation 14, 110.
Puthussery, T., Fletcher, E.L., 2004. Synaptic localization of P2X7 receptors in the rat
retina. J. Comp. Neurol. 472, 1323.
Quigley, H., Addicks, Em, Green, Wr, Maumenee, Abugbuye, 1981. Optic nerve damage
in human glaucoma. II. The site of injury and susceptibility to damage. Arch.
Ophthalmol. 99, 635649.
Quigley, H.A., Addicks, E.M., 1981. Regional differences in the structure of the lamina
cribrosa and their relation to glaucomatous optic nerve damage. Arch. Ophthalmol.
99, 137143.
Quigley, H.A., West, S.K., Rodriguez, J., Munoz, B., Klein, R., Snyder, R., 2001. The
prevalence of glaucoma in a population-based study of Hispanic subjects: proyecto
VER. Arch. Ophthalmol. 119, 18191826.
Quintero, H., Shiga, Y., Belforte, N., Alarcon-Martinez, L., El Hajji, S., Villafranca-
Baughman, D., Dotigny, F., Di Polo, A., 2022. Restoration of mitochondria axonal
transport by adaptor Disc1 supplementation prevents neurodegeneration and rescues
visual function. Cell Rep. 40, 111324.
Radius, R.L., Anderson, D.R., 1980. Breakdown of the normal optic nerve head blood-
brain barrier following acute elevation of intraocular pressure in experimental
animals. Invest. Ophthalmol. Vis. Sci. 19, 244255.
Ramadan, F.M., Upchurch Jr., G.R., Keagy, B.A., Johnson Jr., G., 1990. Endothelial cell
thromboxane production and its inhibition by a calcium-channel blocker. Ann.
Thorac. Surg. 49, 916919.
Rao, H.L., Pradhan, Z.S., Suh, M.H., Moghimi, S., Mansouri, K., Weinreb, R.N., 2020.
Optical coherence tomography angiography in glaucoma. J. Glaucoma 29, 312321.
Rao, H.L., Pradhan, Z.S., Weinreb, R.N., Reddy, H.B., Riyazuddin, M., Dasari, S.,
Palakurthy, M., Puttaiah, N.K., Rao, D.A., Webers, C.A., 2016. Regional comparisons
of optical coherence tomography angiography vessel density in primary open-angle
glaucoma. Am. J. Ophthalmol. 171, 7583.
Rao, H.L., Pradhan, Z.S., Weinreb, R.N., Riyazuddin, M., Dasari, S., Venugopal, J.P.,
Puttaiah, N.K., Rao, D.A., Devi, S., Mansouri, K., Webers, C.A., 2017. A comparison
of the diagnostic ability of vessel density and structural measurements of optical
coherence tomography in primary open angle glaucoma. PLoS One 12, e0173930.
Rattner, A., Nathans, J., 2005. The genomic response to retinal disease and injury:
evidence for endothelin signaling from photoreceptors to glia. J. Neurosci. 25,
45404549.
Rattner, A., Yu, H., Williams, J., Smallwood, P.M., Nathans, J., 2013. Endothelin-2
signaling in the neural retina promotes the endothelial tip cell state and inhibits
angiogenesis. Proc. Natl. Acad. Sci. U. S. A. 110, E3830E3839.
Reese, T.S., Karnovsky, M.J., 1967. Fine structural localization of a blood-brain barrier to
exogenous peroxidase. J. Cell Biol. 34, 207217.
Resch, H., Garhofer, G., Fuchsj¨
ager-Mayrl, G., Hommer, A., Schmetterer, L., 2009.
Endothelial dysfunction in glaucoma. Acta Ophthalmol. 87, 412.
Risner, M.L., Mcgrady, N.R., Boal, A.M., Pasini, S., Calkins, D.J., 2020. TRPV1 supports
axogenic enhanced excitability in response to neurodegenerative stress. Front. Cell.
Neurosci. 14, 603419.
Riva, C.E., Grunwald, J.E., Petrig, B.L., 1986a. Autoregulation of human retinal blood
ow. An investigation with laser Doppler velocimetry. Invest. Ophthalmol. Vis. Sci.
27, 17061712.
Riva, C.E., Grunwald, J.E., Sinclair, S.H., 1983. Laser Doppler Velocimetry study of the
effect of pure oxygen breathing on retinal blood ow. Invest. Ophthalmol. Vis. Sci.
24, 4751.
Riva, C.E., Hero, M., Titze, P., Petrig, B., 1997. Autoregulation of human optic nerve
head blood ow in response to acute changes in ocular perfusion pressure. Graefes
Arch. Clin. Exp. Ophthalmol. 235, 618626.
Riva, C.E., Logean, E., Falsini, B., 2005. Visually evoked hemodynamical response and
assessment of neurovascular coupling in the optic nerve and retina. Prog. Retin. Eye
Res. 24, 183215.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
23
Riva, C.E., Pournaras, C.J., Tsacopoulos, M., 1986b. Regulation of local oxygen tension
and blood ow in the inner retina during hyperoxia. J. Appl. Physiol. 61, 592598.
Riva, C.E., Salgarello, T., Logean, E., Colotto, A., Galan, E.M., Falsini, B., 2004. Flicker-
evoked response measured at the optic disc rim is reduced in ocular hypertension
and early glaucoma. Invest. Ophthalmol. Vis. Sci. 45, 36623668.
Rodríguez, A.M., Rodríguez, J., Giambartolomei, G.H., 2022. Microglia at the crossroads
of pathogen-induced neuroinammation, 17590914221104566 ASN Neuro 14.
Rosengarten, B., Hecht, M., Kaps, M., 2007. Brain activity affects dynamic but not static
autoregulation. Exp. Neurol. 205, 201206.
Rouget, C., 1873. M´
emoire sur le d´
eveloppement, la structure et les propriet´
es
physiologiques des capillaires sanguins et lymphatiques. Arch. Physiol. Norm.
Pathol. 5, 603663.
Roy, C.S., Sherrington, C.S., 1890. On the regulation of the blood-supply of the brain.
J. Physiol. 11, 85158, 17.
Rungta, R.L., Bernier, L.-P., Dissing-Olesen, L., Groten, C.J., Ledue, J.M., Ko, R.,
Drissler, S., Macvicar, B.A., 2016. Ca2+transients in astrocyte ne processes occur
via Ca2+inux in the adult mouse hippocampus. Glia 64, 20932103.
Ryskamp, D.A., Witkovsky, P., Barabas, P., Huang, W., Koehler, C., Akimov, N.P., Lee, S.
H., Chauhan, S., Xing, W., Rentería, R.C., Liedtke, W., Krizaj, D., 2011. The
polymodal ion channel transient receptor potential vanilloid 4 modulates calcium
ux, spiking rate, and apoptosis of mouse retinal ganglion cells. J. Neurosci. 31,
70897101.
Sakagami, K., Kawamura, H., Wu, D.M., Puro, D.G., 2001. Nitric oxide/cGMP-induced
inhibition of calcium and chloride currents in retinal pericytes. Microvasc. Res. 62,
196203.
Sakurai, T., Yanagisawa, M., Takuwa, Y., Miyazaki, H., Kimura, S., Goto, K., Masaki, T.,
1990. Cloning of a cDNA encoding a non-isopeptide-selective subtype of the
endothelin receptor. Nature 348, 732735.
Sancho, M., Klug, N.R., Mughal, A., Koide, M., Huerta De La Cruz, S., Heppner, T.J.,
Bonev, A.D., Hill-Eubanks, D., Nelson, M.T., 2022a. Adenosine signaling activates
ATP-sensitive K(+) channels in endothelial cells and pericytes in CNS capillaries. Sci.
Signal. 15, eabl5405.
Sancho, M., Klug, N.R., Mughal, A., Koide, M., Huerta De La Cruz, S., Heppner, T.J.,
Bonev, A.D., Hill-Eubanks, D., Nelson, M.T., 2022b. Adenosine signaling activates
ATP-sensitive K+channels in endothelial cells and pericytes in CNS capillaries. Sci.
Signal. 15, eabl5405.
Sasaoka, M., Taniguchi, T., Shimazawa, M., Ishida, N., Shimazaki, A., Hara, H., 2006.
Intravitreal injection of endothelin-1 caused optic nerve damage following to ocular
hypoperfusion in rabbits. Exp. Eye Res. 83, 629637.
Schmidl, D., Garhofer, G., Schmetterer, L., 2011. The complex interaction between
ocular perfusion pressure and ocular blood ow - relevance for glaucoma. Exp. Eye
Res. 93, 141155.
Schwartz, B., 1994. Circulatory defects of the optic disk and retina in ocular hypertension
and high pressure open-angle glaucoma. Surv. Ophthalmol. 38 (Suppl. l), S23S34.
Schwartz, B., Rieser, J.C., Fishbein, S.L., 1977. Fluorescein angiographic defects of the
optic disc in glaucoma. Arch. Ophthalmol. 95, 19611974.
Scoles, D., Gray, D.C., Hunter, J.J., Wolfe, R., Gee, B.P., Geng, Y., Masella, B.D., Libby, R.
T., Russell, S., Williams, D.R., Merigan, W.H., 2009. In-vivo imaging of retinal nerve
ber layer vasculature: imaging histology comparison. BMC Ophthalmol. 9, 9.
Sherer, N.M., 2013. Long-distance relationships: do membrane nanotubes regulate cell-
cell communication and disease progression? Mol. Biol. Cell 24, 10951098.
Shiga, Y., Aizawa, N., Tsuda, S., Yokoyama, Y., Omodaka, K., Kunikata, H., Yasui, T.,
Kato, K., Kurashima, H., Miyamoto, E., Hashimoto, M., Nakazawa, T., 2018.
Preperimetric glaucoma prospective study (PPGPS): predicting visual eld
progression with basal optic nerve head blood ow in normotensive PPG eyes.
Transl. Vis. Sci. Technol. 7, 11.
Shiga, Y., Kunikata, H., Aizawa, N., Kiyota, N., Maiya, Y., Yokoyama, Y., Omodaka, K.,
Takahashi, H., Yasui, T., Kato, K., Iwase, A., Nakazawa, T., 2016. Optic nerve head
blood ow, as measured by laser speckle owgraphy, is signicantly reduced in
preperimetric glaucoma. Curr. Eye Res. 41, 14471453.
Shiga, Y., Nishida, T., Jeoung, J.W., Di Polo, A., Fortune, B., 2023. Optical coherence
tomography and optical coherence tomography angiography: essential tools for
detecting glaucoma and disease progression. Front. Ophthalmol. 3 (in press).
Shiga, Y., Omodaka, K., Kunikata, H., Ryu, M., Yokoyama, Y., Tsuda, S., Asano, T.,
Maekawa, S., Maruyama, K., Nakazawa, T., 2013. Waveform analysis of ocular blood
ow and the early detection of normal tension glaucoma. Invest. Ophthalmol. Vis.
Sci. 54, 76997706.
Shiga, Y., Sato, M., Maruyama, K., Takayama, S., Omodaka, K., Himori, N., Kunikata, H.,
Nakazawa, T., 2015. Assessment of short-term changes in optic nerve head
hemodynamics in hyperoxic conditions with laser speckle owgraphy. Curr. Eye Res.
40, 10551062.
Shin, J.D., Wolf, A.T., Harris, A., Verticchio Vercellin, A., Siesky, B., Rowe, L.W.,
Packles, M., Oddone, F., 2022. Vascular biomarkers from optical coherence
tomography angiography and glaucoma: where do we stand in 2021? Acta
Ophthalmol. 100, e377e385.
Shin, J.W., Lee, J., Kwon, J., Choi, J., Kook, M.S., 2017. Regional vascular density-visual
eld sensitivity relationship in glaucoma according to disease severity. Br. J.
Ophthalmol. 101, 16661672.
Siegenthaler, J.A., Sohet, F., Daneman, R., 2013. Sealing off the CNS: cellular and
molecular regulation of blood-brain barriergenesis. Curr. Opin. Neurobiol. 23,
10571064.
Sigal, I.A., Ethier, C.R., 2009. Biomechanics of the optic nerve head. Exp. Eye Res. 88,
799807.
Sims, D.E., 1986. The pericyte–a review. Tissue Cell 18, 153174.
Sines, D., Harris, A., Siesky, B., Januleviciene, I., Haine, C.L., Yung, C.W., Catoira, Y.,
Garzozi, H.J., 2007. The response of retrobulbar vasculature to hypercapnia in
primary open-angle glaucoma and ocular hypertension. Ophthalmic Res. 39, 7680.
Singh, S., Dass, R., 1960a. The central artery of the retina. I. Origin and course. Br. J.
Ophthalmol. 44, 193212.
Singh, S., Dass, R., 1960b. The central artery of the retina. II. A study of its distribution
and anastomoses. Br. J. Ophthalmol. 44, 280299.
Sossi, N., Anderson, D.R., 1983. Effect of elevated intraocular pressure on blood ow.
Occurrence in cat optic nerve head studied with iodoantipyrine I 125. Arch.
Ophthalmol. 101, 98101.
Spaeth, G.L., 1975. Fluorescein angiography: its contributions towards understanding the
mechanisms of visual loss in glaucoma. Trans. Am. Ophthalmol. Soc. 73, 491553.
Srienc, A.I., Kurth-Nelson, Z.L., Newman, E.A., 2010. Imaging retinal blood ow with
laser speckle owmetry. Front. Neuroenergetics 2, 128.
Stefansson, E., Wagner, H.G., Seida, M., 1988. Retinal blood ow and its autoregulation
measured by intraocular hydrogen clearance. Exp. Eye Res. 47, 669678.
Su, W.W., Cheng, S.T., Hsu, T.S., Ho, W.J., 2006. Abnormal ow-mediated vasodilation
in normal-tension glaucoma using a noninvasive determination for peripheral
endothelial dysfunction. Invest. Ophthalmol. Vis. Sci. 47, 33903394.
Suh, M.H., Zangwill, L.M., Manalastas, P.I., Belghith, A., Yarmohammadi, A.,
Medeiros, F.A., Diniz-Filho, A., Saunders, L.J., Weinreb, R.N., 2016. Deep retinal
layer microvasculature dropout detected by the optical coherence tomography
ngiography in glaucoma. Ophthalmology 123, 25092518.
Sun, M.-H., Chen, K.-J., Tsao, Y.-P., Kao, L.-Y., Han, W.-H., Lin, K.-K., Pang, J.-H.S., 2011.
Down-regulation of matrix metalloproteinase-9 by pyrrolidine dithiocarbamate
prevented retinal ganglion cell death after transection of optic nerve in rats. Curr.
Eye Res. 36, 10531063.
Sung, K.R., Lee, S., Park, S.B., Choi, J., Kim, S.T., Yun, S.C., Kang, S.Y., Cho, J.W.,
Kook, M.S., 2009. Twenty-four hour ocular perfusion pressure uctuation and risk of
normal-tension glaucoma progression. Invest. Ophthalmol. Vis. Sci. 50, 52665274.
Sweeney, M., Foldes, G., 2018. It takes two: endothelial-perivascular cell cross-talk in
vascular development and disease. Front Cardiovasc Med 5, 154.
Sweeney, M.D., Zhao, Z., Montagne, A., Nelson, A.R., Zlokovic, B.V., 2019. Blood-brain
barrier: from physiology to disease and back. Physiol. Rev. 99, 2178.
Takihara, Y., Inatani, M., Eto, K., Inoue, T., Kreymerman, A., Miyake, S., Ueno, S.,
Nagaya, M., Nakanishi, A., Iwao, K., Takamura, Y., Sakamoto, H., Satoh, K.,
Kondo, M., Sakamoto, T., Goldberg, J.L., Nabekura, J., Tanihara, H., 2015. In vivo
imaging of axonal transport of mitochondria in the diseased and aged mammalian
CNS. Proc. Natl. Acad. Sci. U. S. A. 112, 1051510520.
Takusagawa, H.L., Liu, L., Ma, K.N., Jia, Y., Gao, S.S., Zhang, M., Edmunds, B.,
Parikh, M., Tehrani, S., Morrison, J.C., Huang, D., 2017. Projection-resolved optical
coherence tomography angiography of macular retinal circulation in glaucoma.
Ophthalmology 124, 15891599.
Talusan, E., Schwartz, B., 1977. Specicity of uorescein angiographic defects of the
optic disc in glaucoma. Arch. Ophthalmol. 95, 21662175.
Tan, Z., Guo, Y., Shrestha, M., Sun, D., Gregory-Ksander, M., Jakobs, T.C., 2022.
Microglia depletion exacerbates retinal ganglion cell loss in a mouse model of
glaucoma. Exp. Eye Res. 225, 109273.
Tarasiuk, O., Scuteri, A., 2022. Role of tunneling nanotubes in the nervous system. Int. J.
Mol. Sci. 23.
Tarasoff-Conway, J.M., Carare, R.O., Osorio, R.S., Glodzik, L., Butler, T., Fieremans, E.,
Axel, L., Rusinek, H., Nicholson, C., Zlokovic, B.V., Frangione, B., Blennow, K.,
M´
enard, J., Zetterberg, H., Wisniewski, T., De Leon, M.J., 2015. Clearance systems in
the brain-implications for Alzheimer disease. Nat. Rev. Neurol. 11, 457470.
Tezel, G., Kass, M.A., Kolker, A.E., Becker, B., Wax, M.B., 1997. Plasma and aqueous
humor endothelin levels in primary open-angle glaucoma. J. Glaucoma 6, 8389.
Thakore, P., Alvarado, M.G., Ali, S., Mughal, A., Pires, P.W., Yamasaki, E., Pritchard, H.
A., Isakson, B.E., Tran, C.H.T., Earley, S., 2021. Brain endothelial cell TRPA1
channels initiate neurovascular coupling. Elife 10, e63040.
Tham, Y.C., Li, X., Wong, T.Y., Quigley, H.A., Aung, T., Cheng, C.Y., 2014. Global
prevalence of glaucoma and projections of glaucoma burden through 2040: a
systematic review and meta-analysis. Ophthalmology 121, 20812090.
Tian, R., Luo, Y., Liu, Q., Cai, M., Li, J., Sun, W., Wang, J., He, C., Liu, Y., Liu, X., 2014.
The effect of claudin-5 overexpression on the interactions of claudin-1 and -2 and
barrier function in retinal cells. Curr. Mol. Med. 14, 12261237.
Tiedt, S., Buchan, A.M., Dichgans, M., Lizasoain, I., Moro, M.A., Lo, E.H., 2022. The
neurovascular unit and systemic biology in stroke implications for translation and
treatment. Nat. Rev. Neurol. 18, 597612.
Tietz, S., Engelhardt, B., 2015. Brain barriers: crosstalk between complex tight junctions
and adherens junctions. J. Cell Biol. 209, 493506.
Tornavaca, O., Chia, M., Dufton, N., Almagro, L.O., Conway, D.E., Randi, A.M.,
Schwartz, M.A., Matter, K., Balda, M.S., 2015. ZO-1 controls endothelial adherens
junctions, cell-cell tension, angiogenesis, and barrier formation. J. Cell Biol. 208,
821838.
Torring, M.S., Aalkjaer, C., Bek, T., 2014. Constriction of porcine retinal arterioles
induced by endothelin-1 and the thromboxane analogue U46619 in vitro decreases
with increasing vascular branching level. Acta Ophthalmol. 92, 232237.
Tribble, J.R., Hui, F., Quintero, H., El Hajji, S., Bell, K., Di Polo, A., Williams, P.A., 2023.
Neuroprotection in glaucoma: mechanisms beyond intraocular pressure lowering.
Mol. Aspect. Med. 92, 101193.
Trost, A., Bruckner, D., Rivera, F.J., Reitsamer, H.A., 2019. Pericytes in the retina. Adv.
Exp. Med. Biol. 1122, 126.
Trost, A., Lange, S., Schroedl, F., Bruckner, D., Motloch, K.A., Bogner, B., Kaser-
Eichberger, A., Strohmaier, C., Runge, C., Aigner, L., Rivera, F.J., Reitsamer, H.A.,
2016. Brain and retinal pericytes: origin, function and role. Front. Cell. Neurosci. 10,
20.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
24
Tsukahara, S., 1978. Hyperpermeable disc capillaries in glaucoma. Adv. Ophthalmol. 35,
6572.
Turowski, P., Martinelli, R., Crawford, R., Wateridge, D., Papageorgiou, A.P.,
Lampugnani, M.G., Gamp, A.C., Vestweber, D., Adamson, P., Dejana, E.,
Greenwood, J., 2008. Phosphorylation of vascular endothelial cadherin controls
lymphocyte emigration. J. Cell Sci. 121, 2937.
Uemura, A., Ogawa, M., Hirashima, M., Fujiwara, T., Koyama, S., Takagi, H., Honda, Y.,
Wiegand, S.J., Yancopoulos, G.D., Nishikawa, S.-I., 2002. Recombinant angiopoietin-
1 restores higher-order architecture of growing blood vessels in mice in the absence
of mural cells. J. Clin. Invest. 110, 16191628.
Uemura, M.T., Maki, T., Ihara, M., Lee, V.M.Y., Trojanowski, J.Q., 2020. Brain
microvascular pericytes in vascular cognitive impairment and dementia. Front.
Aging Neurosci. 12, 80.
Underly, R.G., Levy, M., Hartmann, D.A., Grant, R.I., Watson, A.N., Shih, A.Y., 2017.
Pericytes as inducers of rapid, matrix metalloproteinase-9-dependent capillary
damage during ischemia. J. Neurosci. 37, 129140.
Ushiwata, I., Ushiki, T., 1990. Cytoarchitecture of the smooth muscles and pericytes of
rat cerebral blood vessels. A scanning electron microscopic study. J. Neurosurg. 73,
8290.
Van Beek, A.H., Claassen, J.A., Rikkert, M.G., Jansen, R.W., 2008. Cerebral
autoregulation: an overview of current concepts and methodology with special focus
on the elderly. J. Cerebr. Blood Flow Metabol. 28, 10711085.
Vanhoutte, P.M., Tang, E.H., 2008. Endothelium-dependent contractions: when a good
guy turns bad. J. Physiol. 586, 52955304.
Vanlandewijck, M., He, L., M¨
ae, M.A., Andrae, J., Ando, K., Del Gaudio, F., Nahar, K.,
Lebouvier, T., Lavi˜
na, B., Gouveia, L., Sun, Y., Raschperger, E., R¨
as¨
anen, M., Zarb, Y.,
Mochizuki, N., Keller, A., Lendahl, U., Betsholtz, C., 2018. A molecular atlas of cell
types and zonation in the brain vasculature. Nature 554, 475480.
Venkataraman, S.T., Flanagan, J.G., Hudson, C., 2010. Vascular reactivity of optic nerve
head and retinal blood vessels in glaucoma. Microcirculation 17, 568581.
Vohra, R., Aldana, B.I., Bulli, G., Skytt, D.M., Waagepetersen, H., Bergersen, L.H.,
Kolko, M., 2019. Lactate-mediated protection of retinal ganglion cells. J. Mol. Biol.
431, 18781888.
Vorbrodt, A.W., Dobrogowska, D.H., 2003. Molecular anatomy of intercellular junctions
in brain endothelial and epithelial barriers: electron microscopists view. Brain Res.
Rev. 42, 221242.
Wan, K.H., Lam, A.K.N., Leung, C.K., 2018. Optical coherence tomography angiography
compared with optical coherence tomography macular measurements for detection
of glaucoma. JAMA Ophthalmol. 136, 866874.
Wang, L., Burgoyne, C.F., Cull, G., Thompson, S., Fortune, B., 2014a. Static blood ow
autoregulation in the optic nerve head in normal and experimental glaucoma. Invest.
Ophthalmol. Vis. Sci. 55, 873880.
Wang, L., Cull, G., Burgoyne, C.F., Thompson, S., Fortune, B., 2014b. Longitudinal
alterations in the dynamic autoregulation of optic nerve head blood ow revealed in
experimental glaucoma. Invest. Ophthalmol. Vis. Sci. 55, 35093516.
Wang, X., Veruki, M.L., Bukoreshtliev, N.V., Hartveit, E., Gerdes, H.-H., 2010. Animal
cells connected by nanotubes can be electrically coupled through interposed gap-
junction channels. Proc. Natl. Acad. Sci. U. S. A. 107, 1719417199.
Wang, X., Zhao, L., Zhang, J., Fariss, R.N., Ma, W., Kretschmer, F., Wang, M., Qian, H.H.,
Badea, T.C., Diamond, J.S., Gan, W.-B., Roger, J.E., Wong, W.T., 2016. Requirement
for microglia for the maintenance of synaptic function and integrity in the mature
retina. J. Neurosci. 36, 28272842.
Wareham, L.K., Calkins, D.J., 2020. The neurovascular unit in glaucomatous
neurodegeneration. Front. Cell Dev. Biol. 8, 452.
Wareham, L.K., Liddelow, S.A., Temple, S., Benowitz, L.I., Di Polo, A., Wellington, C.,
Goldberg, J.L., He, Z., Duan, X., Bu, G., Davis, A.A., Shekhar, K., Torre, A., Chan, D.
C., Canto-Soler, M.V., Flanagan, J.G., Subramanian, P., Rossi, S., Brunner, T.,
Bovenkamp, D.E., Calkins, D.J., 2022. Solving neurodegeneration: common
mechanisms and strategies for new treatments. Mol. Neurodegener. 17, 23.
Watson, E.C., Grant, Z.L., Coultas, L., 2017. Endothelial cell apoptosis in angiogenesis
and vessel regression. Cell. Mol. Life Sci. 74, 43874403.
Wei, H.S., Kang, H., Rasheed, I.D., Zhou, S., Lou, N., Gershteyn, A., Mcconnell, E.D.,
Wang, Y., Richardson, K.E., Palmer, A.F., Xu, C., Wan, J., Nedergaard, M., 2016a.
Erythrocytes are oxygen-sensing regulators of the cerebral microcirculation. Neuron
91, 851862.
Wei, L., Sheng, H., Chen, L., Hao, B., Shi, X., Chen, Y., 2016b. Effect of pannexin-1 on the
release of glutamate and cytokines in astrocytes. J. Clin. Neurosci. 23, 135141.
Weidmann, S., 1969. Electrical coupling between myocardial cells. Prog. Brain Res. 31,
275281.
Weir, N., Longden, T.A., 2021. Pathologically entangled: brain trauma-evoked ROS
imbalance disrupts Kir channel function in distant peripheral vessels. Function 2,
zqab021.
Wessel, F., Winderlich, M., Holm, M., Frye, M., Rivera-Galdos, R., Vockel, M.,
Linnepe, R., Ipe, U., Stadtmann, A., Zarbock, A., Nottebaum, A.F., Vestweber, D.,
2014. Leukocyte extravasation and vascular permeability are each controlled in vivo
by different tyrosine residues of VE-cadherin. Nat. Immunol. 15, 223230.
Westheimer, G., 2007. The ON-OFF dichotomy in visual processing: from receptors to
perception. Prog. Retin. Eye Res. 26, 636648.
Wilkison, S.J., Bright, C.L., Vancini, R., Song, D.J., Bomze, H.M., Cartoni, R., 2021. Local
accumulation of axonal mitochondria in the optic nerve glial lamina precedes
myelination. Front. Neuroanat. 15, 678501-678501.
Willebrords, J., Crespo Yanguas, S., Maes, M., Decrock, E., Wang, N., Leybaert, L.,
Kwak, B.R., Green, C.R., Cogliati, B., Vinken, M., 2016. Connexins and their channels
in inammation. Crit. Rev. Biochem. Mol. Biol. 51, 413439.
Williams, P., Harder, J., John, S.W.M., 2017a. Glaucoma as a metabolic optic
neuropathy: making the case for nicotinamide treatment in glaucoma. J. Glaucoma
26, 11611168.
Williams, P.A., Harder, J.M., Foxworth, N.E., Cochran, K.E., Philip, V.M., Porciatti, V.,
Smithies, O., John, S.W.M., 2017b. Vitamin B(3) modulates mitochondrial
vulnerability and prevents glaucoma in aged mice. Science 355, 756760.
Wright, W.S., Harris, N.R., 2008. Ozagrel attenuates early streptozotocin-induced
constriction of arterioles in the mouse retina. Exp. Eye Res. 86, 528536.
Yabana, T., Shiga, Y., Kawasaki, R., Omodaka, K., Takahashi, H., Kimura, K., Togashi, K.,
Horii, T., Sasaki, K., Yuasa, T., Nakazawa, T., 2017. Evaluating retinal vessel
diameter with optical coherence tomography in normal-tension glaucoma patients.
Jpn. J. Ophthalmol. 61, 378387.
Yamashita, Y.M., Inaba, M., Buszczak, M., 2018. Specialized intercellular
communications via cytonemes and nanotubes. Annu. Rev. Cell Dev. Biol. 34, 5984.
Yamazaki, Y., Kanekiyo, T., 2017. Blood-brain barrier dysfunction and the pathogenesis
of Alzheimers disease. Int. J. Mol. Sci. 18.
Yanagisawa, M., Kurihara, H., Kimura, S., Tomobe, Y., Kobayashi, M., Mitsui, Y.,
Yazaki, Y., Goto, K., Masaki, T., 1988. A novel potent vasoconstrictor peptide
produced by vascular endothelial cells. Nature 332, 411415.
Yang, A.C., Vest, R.T., Kern, F., Lee, D.P., Agam, M., Maat, C.A., Losada, P.M., Chen, M.
B., Schaum, N., Khoury, N., Toland, A., Calcuttawala, K., Shin, H., P´
alovics, R.,
Shin, A., Wang, E.Y., Luo, J., Gate, D., Schulz-Schaeffer, W.J., Chu, P.,
Siegenthaler, J.A., Mcnerney, M.W., Keller, A., Wyss-Coray, T., 2022. A human brain
vascular atlas reveals diverse mediators of Alzheimers risk. Nature 603, 885892.
Yang, D., Chen, M., Yang, S., Deng, F., Guo, X., 2023. Connexin hemichannels and
pannexin channels in toxicity: recent advances and mechanistic insights. Toxicology
488, 153488.
Yang, G., Zhang, Y., Ross, M.E., Iadecola, C., 2003. Attenuation of activity-induced
increases in cerebellar blood ow in mice lacking neuronal nitric oxide synthase.
Am. J. Physiol. Heart Circ. Physiol. 285, H298H304.
Yao, Y., Chen, Z.L., Norris, E.H., Strickland, S., 2014. Astrocytic laminin regulates
pericyte differentiation and maintains blood brain barrier integrity. Nat. Commun. 5,
3413.
Yarmohammadi, A., Zangwill, L.M., Diniz-Filho, A., Suh, M.H., Manalastas, P.I.,
Fatehee, N., Youse, S., Belghith, A., Saunders, L.J., Medeiros, F.A., Huang, D.,
Weinreb, R.N., 2016. Optical coherence tomography angiography vessel density in
healthy, glaucoma suspect, and glaucoma eyes. Invest. Ophthalmol. Vis. Sci. 57,
Oct451O459.
Yazdankhah, M., Shang, P., Ghosh, S., Hose, S., Liu, H., Weiss, J., Fitting, C.S., Bhutto, I.
A., Zigler Jr., J.S., Qian, J., Sahel, J.A., Sinha, D., Stepicheva, N.A., 2021. Role of glia
in optic nerve. Prog. Retin. Eye Res. 81, 100886.
Ye, X.D., Laties, A.M., Stone, R.A., 1990. Peptidergic innervation of the retinal
vasculature and optic nerve head. Invest. Ophthalmol. Vis. Sci. 31, 17311737.
Yemisci, M., Gursoy-Ozdemir, Y., Vural, A., Can, A., Topalkara, K., Dalkara, T., 2009.
Pericyte contraction induced by oxidative-nitrative stress impairs capillary reow
despite successful opening of an occluded cerebral artery. Nat. Med. 15, 10311037.
Yokoyama, U., Iwatsubo, K., Umemura, M., Fujita, T., Ishikawa, Y., 2013. The prostanoid
EP4 receptor and its signaling pathway. Pharmacol. Rev. 65, 10101052.
Yu, D.-Y., Cringle, S.J., Yu, P.K., Balaratnasingam, C., Mehnert, A., Sarunic, M.V., An, D.,
Su, E.-N., 2019. Retinal capillary perfusion: spatial and temporal heterogeneity.
Prog. Retin. Eye Res. 70, 2354.
Zambach, S.A., Cai, C., Helms, H.C.C., Hald, B.O., Dong, Y., Fordsmann, J.C., Nielsen, R.
M., Hu, J., Lønstrup, M., Brodin, B., Lauritzen, M.J., 2021. Precapillary sphincters
and pericytes at rst-order capillaries as key regulators for brain capillary perfusion.
Proc. Natl. Acad. Sci. U. S. A. 118.
Zhang, T., Wu, D.M., Xu, G.-Z., Puro, D.G., 2011. The electrotonic architecture of the
retinal microvasculature: modulation by angiotensin II. J. Physiol. 589, 23832399.
Zhang, W., Davis, C.M., Zeppenfeld, D.M., Golgotiu, K., Wang, M.X., Haveliwala, M.,
Hong, D., Li, Y., Wang, R.K., Iliff, J.J., Alkayed, N.J., 2021. Role of endothelium-
pericyte signaling in capillary blood ow response to neuronal activity. J. Cerebr.
Blood Flow Metabol. 41, 18731885.
Zhang, X., Cheng, M., Chintala, S.K., 2004a. Kainic acid-mediated upregulation of matrix
metalloproteinase-9 promotes retinal degeneration. Invest. Ophthalmol. Vis. Sci. 45,
23742383.
Zhang, X., Cheng, M., Chintala, S.K., 2004b. Optic nerve ligation leads to astrocyte-
associated matrix metalloproteinase-9 induction in the mouse retina. Neurosci. Lett.
356, 140144.
Zhang, X., Chintala, S.K., 2004. Inuence of interleukin-1 beta induction and mitogen-
activated protein kinase phosphorylation on optic nerve ligation-induced matrix
metalloproteinase-9 activation in the retina. Exp. Eye Res. 78, 849860.
Zhang, Y., Chen, K., Sloan, S.A., Bennett, M.L., Scholze, A.R., Okeeffe, S., Phatnani, H.P.,
Guarnieri, P., Caneda, C., Ruderisch, N., Deng, S., Liddelow, S.A., Zhang, C.,
Daneman, R., Maniatis, T., Barres, B.A., Wu, J.Q., 2014. An RNA-sequencing
transcriptome and splicing database of glia, neurons, and vascular cells of the
cerebral cortex. J. Neurosci. 34, 1192911947.
Zhao, W., Wang, R., 2002. H(2)S-induced vasorelaxation and underlying cellular and
molecular mechanisms. Am. J. Physiol. Heart Circ. Physiol. 283, H474H480.
Zhao, Y., Vanhoutte, P.M., Leung, S.W., 2015a. Vascular nitric oxide: beyond eNOS.
J. Pharmacol. Sci. 129, 8394.
Zhao, Z., Nelson, A.R., Betsholtz, C., Zlokovic, B.V., 2015b. Establishment and
dysfunction of the blood-brain barrier. Cell 163, 10641078.
Zhao, Z., Yu, X., Yang, X., Zhang, J., Zhang, D., Sun, N., Fan, Z., 2020. Elevated
intraocular pressure causes cellular and molecular retinal injuries, advocating a
more moderate intraocular pressure setting during phacoemulsication surgery. Int.
Ophthalmol. 40, 33233336.
L. Alarcon-Martinez et al.
Progress in Retinal and Eye Research 97 (2023) 101217
25
Zhu, M., Ackerman, J.J., Sukstanskii, A.L., Yablonskiy, D.A., 2006. How the body
controls brain temperature: the temperature shielding effect of cerebral blood ow.
J. Appl. Physiol. 101, 14811488.
Zlokovic, B.V., 2011. Neurovascular pathways to neurodegeneration in Alzheimers
disease and other disorders. Nat. Rev. Neurosci. 12, 723738.
Zonta, M., Angulo, M.C., Gobbo, S., Rosengarten, B., Hossmann, K.A., Pozzan, T.,
Carmignoto, G., 2003. Neuron-to-astrocyte signaling is central to the dynamic
control of brain microcirculation. Nat. Neurosci. 6, 4350.
L. Alarcon-Martinez et al.
... indicates that pericytes play a role in several ocular diseases, such as diabetic retinopathy [10] , choroidal neovascularization [11] , corneal neovascularization [12] , retinopathy of prematurity [13] , and glaucoma [14] . ...
... 138 Increased intraocular pressure in the eye causes damage to IP-TNTs that impairs the blood flow and represents a risk factor for the development of glaucoma. 139,140 An important metabolic aspect of interactions between PCs and ECs was revealed recently. 31 ECs are highly glycolytic and the larger part of lactate that is produced is secreted into the ECM making the lactate available for uptake by PCs. ...
Article
Full-text available
Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX–XXX, XXXX)
... When NVU physiological function is normal, the unit senses regional mechanical stress, neurotransmitters released by neurons, or other types of biological signals, and transmits electrochemical signals to nearby blood vessels or glial cells that trigger the downstream biological responses. The interaction between neurons, glial cells, and blood vessels within the NVU is crucial; any abnormality in these components can disrupt the microenvironment's homeostasis [23]. In this section, we discuss the cellular and molecular mechanisms of the NVU and their pathophysiological implications in glaucoma and how the function is altered with glaucomatous optic neuropathy. ...
Article
Full-text available
The mechanical theory of glaucoma indicates that high intraocular pressure (IOP) leads to glaucomatous optic nerve damage. However, nearly half of primary open‐angle glaucoma patients with normal intraocular pressure also exhibit progression of what appears to be glaucomatous optic nerve damage. Our earlier prospective study identified for the first time that the relatively low intracranial pressure (ICP) is also an important risk factor for progressive glaucomatous injury of normal‐tension glaucoma. When considering the results of studies in nonhuman primates, clinical research, large‐scale natural‐population studies, and basic laboratory investigations, a new understanding of the pathophysiology of glaucoma, the “Dual‐Pressure Theory”, has been proposed. This theory states that “either high IOP or low ICP contributes to increasing the translaminar cribrosa pressure difference; it is the pressure difference rather than the IOP alone that results in the glaucomatous optic neuropathy”. Here, we provide a systematic introduction to Dual‐Pressure Theory relating to glaucoma, the form of a research map, an outline of basic laboratory investigations, the main methodology, and research updates.
Article
Full-text available
Purpose: To investigate the relationship between glaucoma, pseudoexfoliation and hearing loss (HL). Methods: A systematic literature search following PRISMA guidelines was conducted using the PubMed, Embase, Scopus and Cochrane databases from 1995 up to 28 August 2023. Results: Thirty studies out of the 520 records screened met the inclusion criteria and were included. Most articles (n = 20) analysed the association between pseudoexfoliation syndrome (XFS) and HL, showing XFS patients to have higher prevalence of sensorineural hearing loss (SNHL) at both speech frequencies (0.25, 0.5, 1 and 2 kHz), and higher frequencies (4 and 8 kHz) compared to controls in most cases. No significant differences in prevalence or level of HL between XFS and pseudoexfoliative glaucoma (XFG) were detected in most studies. Eight articles analysed the relationship between primary open-angle glaucoma (POAG) and HL. Overall, a positive association between the two conditions was highlighted across all studies except for two cases. Similarly, articles focusing on NTG and HL (n = 4) showed a positive association in most cases. The role of autoimmunity and, in particular, the presence of antiphosphatidylserine antibodies (APSA) in patients with NTG and HL suggested an underlying autoimmune or vascular mechanism contributing to their pathogenesis. Only one study analysed the relationship between angle-closure glaucoma (ACG) and HL, showing higher incidence of ACG in patients with SNHL compared to normal hearing controls. Conclusions: Most studies detected an association between XFS and HL as well as POAG/NTG/ACG and HL, suggesting the presence of a similar pathophysiology of neurodegeneration. However, given the strength of the association of XFS with HL, it remains unclear whether the presence of XFG is further associated with SNHL. Further research specifically targeted to assess the correlation between glaucoma, XFS and HL is warranted to provide a more comprehensive understanding of this association.
Article
Full-text available
Glaucoma is one of the leading causes of irreversible blindness worldwide and vision loss in the disease results from the deterioration of retinal ganglion cells (RGC) and their axons. Metabolic dysfunction of RGC plays a significant role in the onset and progression of the disease in both human patients and rodent models, highlighting the need to better define the mechanisms regulating cellular energy metabolism in glaucoma. This study sought to determine if Sarm1, a gene involved in axonal degeneration and NAD+ metabolism, contributes to glaucomatous RGC loss in a mouse model with chronic elevated intraocular pressure (IOP). Our data demonstrate that after 16 weeks of elevated IOP, Sarm1 knockout (KO) mice retain significantly more RGC than control animals. Sarm1 KO mice also performed significantly better when compared to control mice during optomotor testing, indicating that visual function is preserved in this group. Our findings also indicate that Sarm1 KO mice display mild ocular developmental abnormalities, including reduced optic nerve axon diameter and lower visual acuity than controls. Finally, we present data to indicate that SARM1 expression in the optic nerve is most prominently associated with oligodendrocytes. Taken together, these data suggest that attenuating Sarm1 activity through gene therapy, pharmacologic inhibition, or NAD+ supplementation, may be a novel therapeutic approach for patients with glaucoma.
Article
Full-text available
Early diagnosis and detection of disease progression are critical to successful therapeutic intervention in glaucoma, the leading cause of irreversible blindness worldwide. Optical coherence tomography (OCT) is a non-invasive imaging technique that allows objective quantification in vivo of key glaucomatous structural changes in the retina and the optic nerve head (ONH). Advances in OCT technology have increased the scan speed and enhanced image quality, contributing to early glaucoma diagnosis and monitoring, as well as the visualization of critically important structures deep within the ONH, such as the lamina cribrosa. OCT angiography (OCTA) is a dye-free technique for noninvasively assessing ocular microvasculature, including capillaries within each plexus serving the macula, peripapillary retina and ONH regions, as well as the deeper vessels of the choroid. This layer-specific assessment of the microvasculature has provided evidence that retinal and choroidal vascular impairments can occur during early stages of glaucoma, suggesting that OCTA-derived measurements could be used as biomarkers for enhancing detection of glaucoma and its progression, as well as to reveal novel insights about pathophysiology. Moreover, these innovations have demonstrated that damage to the macula, a critical region for the vision-related quality of life, can be observed in the early stages of glaucomatous eyes, leading to a paradigm shift in glaucoma monitoring. Other advances in software and hardware, such as artificial intelligence-based algorithms, adaptive optics, and visible-light OCT, may further benefit clinical management of glaucoma in the future. This article reviews the utility of OCT and OCTA for glaucoma diagnosis and disease progression detection, emphasizes the importance of detecting macula damage in glaucoma, and highlights the future perspective of OCT and OCTA. We conclude that the OCT and OCTA are essential glaucoma detection and monitoring tools, leading to clinical and economic benefits for patients and society.
Article
Full-text available
Purpose: Tissue stiffening and alterations in retinal blood flow have both been suggested as causative mechanisms of glaucomatous damage. We tested the hypothesis that retinal blood vessels also stiffen, using laser speckle flowgraphy (LSFG) to characterize vascular resistance. Methods: In the longitudinal Portland Progression Project, 231 eyes of 124 subjects received LSFG scans of the optic nerve head (ONH) and automated perimetry every 6 months for six visits. Eyes were classified as either "glaucoma suspect" or "glaucoma" eyes based on the presence of functional loss on the first visit. Vascular resistance was quantified using the mean values of several instrument-defined parameterizations of the pulsatile waveform measured by LSFG, either in major vessels within the ONH (serving the retina) or in capillaries within ONH tissue, and age-adjusted using a separate group of 127 healthy eyes of 63 individuals. Parameters were compared against the severity and rate of change of functional loss using mean deviation (MD) over the six visits, within the two groups. Results: Among 118 "glaucoma suspect" eyes (average MD, -0.4 dB; rate, -0.45 dB/y), higher vascular resistance was related to faster functional loss, but not current severity of loss. Parameters measured in major vessels were stronger predictors of rate than parameters measured in tissue. Among 113 "glaucoma" eyes (average MD, -4.3 dB; rate, -0.53 dB/y), higher vascular resistance was related to more severe current loss but not rate of loss. Conclusions: Higher retinal vascular resistance and, by likely implication, stiffer retinal vessels were associated with more rapid functional loss in eyes without significant existing loss at baseline.
Article
Full-text available
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Article
Full-text available
The blood-brain barrier (BBB) consists of specialized cells that tightly regulate the in- and outflow of molecules from the blood to brain parenchyma, protecting the brain’s microenvironment. If one of the BBB components starts to fail, its dysfunction can lead to a cascade of neuroinflammatory events leading to neuronal dysfunction and degeneration. Preliminary imaging findings suggest that BBB dysfunction could serve as an early diagnostic and prognostic biomarker for a number of neurological diseases. This review aims to provide clinicians with an overview of the emerging field of BBB imaging in humans by answering three key questions: (1. Disease) In which diseases could BBB imaging be useful? (2. Device) What are currently available imaging methods for evaluating BBB integrity? And (3. Distribution) what is the potential of BBB imaging in different environments, particularly in resource limited settings? We conclude that further advances are needed, such as the validation, standardization and implementation of readily available, low-cost and non-contrast BBB imaging techniques, for BBB imaging to be a useful clinical biomarker in both resource-limited and well-resourced settings.
Article
Full-text available
Pericytes are a heterogeneous population of mesenchymal cells located on the abluminal surface of microvessels, where they provide structural and biochemical support. Pericytes have been implicated in numerous lung diseases including pulmonary arterial hypertension (PAH) and allergic asthma due to their ability to differentiate into scar-forming myofibroblasts, leading to collagen deposition and matrix remodelling and thus driving tissue fibrosis. Pericyte-extracellular matrix interactions as well as other biochemical cues play crucial roles in these processes. In this review, we give an overview of lung pericytes, the key pro-fibrotic mediators they interact with, and detail recent advances in preclinical studies on how pericytes are disrupted and contribute to lung diseases including PAH, allergic asthma, and chronic obstructive pulmonary disease (COPD). Several recent studies using mouse models of PAH have demonstrated that pericytes contribute to these pathological events; efforts are currently underway to mitigate pericyte dysfunction in PAH by targeting the TGF-β, CXCR7, and CXCR4 signalling pathways. In allergic asthma, the dissociation of pericytes from the endothelium of blood vessels and their migration towards inflamed areas of the airway contribute to the characteristic airway remodelling observed in allergic asthma. Although several factors have been suggested to influence this migration such as TGF-β, IL-4, IL-13, and periostin, recent evidence points to the CXCL12/CXCR4 pathway as a potential therapeutic target. Pericytes might also play an essential role in lung dysfunction in response to ageing, as they are responsive to environmental risk factors such as cigarette smoke and air pollutants, which are the main drivers of COPD. However, there is currently no direct evidence delineating the contribution of pericytes to COPD pathology. Although there is a lack of human clinical data, the recent available evidence derived from in vitro and animal-based models shows that pericytes play important roles in the initiation and maintenance of chronic lung diseases and are amenable to pharmacological interventions. Therefore, further studies in this field are required to elucidate if targeting pericytes can treat lung diseases.
Article
Full-text available
Pericytes, attached to the surface of capillaries, play an important role in regulating local blood flow. Using optogenetic tools and genetically encoded reporters in conjunction with confocal and multiphoton imaging techniques, the 3D structure, anatomical organization, and physiology of pericytes have recently been the subject of detailed examination. This work has revealed novel functions of pericytes and morphological features such as tunneling nanotubes in brain and tunneling microtubes in heart. Here, we discuss the state of our current understanding of the roles of pericytes in blood flow control in brain and heart, where functions may differ due to the distinct spatiotemporal metabolic requirements of these tissues. We also outline the novel concept of electro-metabolic signaling, a universal mechanistic framework that links tissue metabolic state with blood flow regulation by pericytes and vascular smooth muscle cells, with capillary KATP and Kir2.1 channels as primary sensors. Finally, we present major unresolved questions and outline how they can be addressed.
Article
Full-text available
Microglia are dynamic guardians of neural tissue and the resident immune cells of the central nervous system (CNS). The disease-associated microglial signature (DAM), also known as the microglial neurodegenerative phenotype (MGnD), has gained significant attention in recent years as a fundamental microglial response common to various neurodegenerative disease pathologies. Interestingly, this signature shares many features in common with developmental microglia, suggesting the existence of recycled gene programs which play a role both in early neural circuit formation as well as in response to aging and disease. In addition, recent advances in single cell RNA sequencing have revealed significant heterogeneity within the original DAM signature, with contributions from both yolk sac-derived microglia as well as bone marrow-derived macrophages. In this review, we examine the role of the DAM signature in retinal development and disease, highlighting crosstalk between resident microglia and infiltrating monocytes which may critically contribute to the underlying mechanisms of age-related neurodegeneration.
Article
Full-text available
Despite the abundance of capillary thin-strand pericytes and their proximity to neurons and glia, little is known of the contributions of these cells to the control of brain hemodynamics. We demonstrate that the pharmacological activation of thin-strand pericyte KATP channels profoundly hyperpolarizes these cells, dilates upstream penetrating arterioles and arteriole-proximate capillaries, and increases capillary blood flow. Focal stimulation of pericytes with a KATP channel agonist is sufficient to evoke this response, mediated via KIR2.1 channel-dependent retrograde propagation of hyperpolarizing signals, whereas genetic inactivation of pericyte KATP channels eliminates these effects. Critically, we show that decreasing extracellular glucose to less than 1 mM or inhibiting glucose uptake by blocking GLUT1 transporters in vivo flips a mechanistic energy switch driving rapid KATP-mediated pericyte hyperpolarization to increase local blood flow. Together, our findings recast capillary pericytes as metabolic sentinels that respond to local energy deficits by increasing blood flow to neurons to prevent energetic shortfalls.
Article
The neurovascular unit (NVU) plays an essential role in regulating neurovascular coupling, which refers to the communication between neurons, glia, and vascular cells to control the supply of oxygen and nutrients in response to neural activity. Cellular elements of the NVU coordinate to establish an anatomical barrier to separate the central nervous system from the milieu of the periphery system, restricting the free movement of substances from the blood to the brain parenchyma and maintaining central nervous system homeostasis. In Alzheimer's disease, amyloid-β deposition impairs the normal functions of NVU cellular elements, thus accelerating the disease progression. Here, we aim to describe the current knowledge of the NVU cellular elements, including endothelial cells, pericytes, astrocytes, and microglia, in regulating the blood-brain barrier integrity and functions in physiology as well as alterations encountered in Alzheimer's disease. Furthermore, the NVU functions as a whole, therefore specific labeling and targeting NVU components in vivo enable us to understand the mechanism mediating cellular communication. We review approaches including commonly used fluorescent dyes, genetic mouse models, and adeno-associated virus vectors for imaging and targeting NVU cellular elements in vivo.
Article
Connexin hemichannels and pannexin channels are two types of transmembrane channels that allow autocrine/paracrine signalling through the exchange of ions and molecules between the intra- and extracellular compartments. However, owing to the poor selectivity of permeable ions and metabolites, the massive opening of these plasma membrane channels can lead to an excessive influx of toxic substances and an outflux of essential metabolites, such as adenosine triphosphate, glutathione, glutamate and ions, resulting in unbalanced cell homeostasis and impaired cell function. It is becoming increasingly clear that these channels can be activated in response to external stimuli and are involved in toxicity, yet their concrete mechanistic roles in the toxic effects induced by stress and various environmental changes remain poorly defined. This review provides an updated understanding of connexin hemichannels and pannexin channels in response to multiple extrinsic stressors and how these activated channels and their permeable messengers participate in toxicological pathways and processes, including inflammation, oxidative damage, intracellular calcium imbalance, bystander DNA damage and excitotoxicity.