ArticlePDF AvailableLiterature Review

Abstract

Background: Cytoskeleton is one of the essential forms of protein, important in the existence of both eukaryotic as well as prokaryotic cells. Its transformation plays a vital role in cell division and intracellular transportation by facilitating intracellular vesicular traffic. Among the various tissue types in the body, the neural tissue exhibits the maximum heterogeneity, and hence the role of cytoskeleton at both developmental and functional levels becomes paramount. Cytoskeleton dynamics have been established in the neural physiology, but only at the level of axonal development and growth. Retina has not been adequately studied in the context of cytoskeletal proteins. Methods: We reviewed the last 10 years of literature with reference to the development, growth, degeneration, and regeneration of the retina and the role of cytoskeleton in each aspect. We have focused on various changes that the retina undergoes at the cytosolic and cytoskeletal levels in the course of degeneration as well as regeneration. Findings: For this review, we compiled research articles pertaining to the role of cytoskeletal and other associated proteins involved in development of retina, which used various animal models. The effect of SNPs in the cytoskeletal proteins and their impact in retinal degeneration is also discussed. Conclusion: Studies describing the role of cytoskeleton in the anatomy and physiology of retina and its layers, although they are few, collectively provide an opportunity to understand retinal development in the context of cytoskeleton dynamics.
Critical ReviewsTM in Eukaryotic Gene Expression, 24(3):255-268 (2014)
255
1045-4403/14/$35.00 © 2014 Begell House, Inc. www.begellhouse.com
Cytoskeleton Dynamics in the Retina
Akshay Anand1,*,** Sridhar Bammidi1,** & Parul Bali2
1Department of Neurology, PGIMER, Chandigarh 160012, India; 2Department of Biophysics, Panjab University, Chan-
digarh 160014, India
* Address all correspondence to: Akshay Anand, Associate Professor, Neuroscience Research Lab, Department of Neurology, PGIMER, Chan-
digarh- 16001; Tel.: +91-991429090; Email: akshay1anand@rediffmail.com.
**Both are rst authors with equal contribution.
ABSTRACT: Background: Cytoskeleton is one of the essential forms of protein, important in the existence of
both eukaryotic as well as prokaryotic cells. Its transformation plays a vital role in cell division and intracellular
transportation by facilitating intracellular vesicular trafc. Among the various tissue types in the body, the neural
tissue exhibits the maximum heterogeneity, and hence the role of cytoskeleton at both developmental and func-
tional levels becomes paramount. Cytoskeleton dynamics have been established in the neural physiology, but only
at the level of axonal development and growth. Retina has not been adequately studied in the context of cytoskel-
etal proteins. Methods: We reviewed the last 10 years of literature with reference to the development, growth, de-
generation, and regeneration of the retina and the role of cytoskeleton in each aspect. We have focused on vari-
ous changes that the retina undergoes at the cytosolic and cytoskeletal levels in the course of degeneration as well
as regeneration. Findings: For this review, we compiled research articles pertaining to the role of cytoskeletal
and other associated proteins involved in development of retina, which used various animal models. The effect
of SNPs in the cytoskeletal proteins and their impact in retinal degeneration is also discussed. Conclusion: Stud-
ies describing the role of cytoskeleton in the anatomy and physiology of retina and its layers, although they are
few, collectively provide an opportunity to understand retinal development in the context of cytoskeleton dynamics.
KEY WORDS: microtubule, intermediate lament, microlament, cytoskeleton
I. INTRODUCTION
The anatomical, physiological, and functional as-
pects of life start from a single cell and can be best
understood through study at the cellular level. The
zygote, from the time of fertilization, undergoes
a series of divisions, physiological and structural
changes, and a vast array of differentiation and
compartmentalization at both inter- and intracellular
levels to reach its fate. Furthermore, these cellular
mechanisms occur in a series of well-dened and
accurate mechanisms of transcription and transla-
tion of protein macromolecules that govern physi-
ological and phenotypic expression. The cytosol
stores various codes for its behavior and metabolic
activities such as migration, translocation, nutrition,
phagocytosis, division, differentiation, and signal-
ing. Codes in the form of proteins can express struc-
turally as well as functionally. One of the essential
forms of proteins that is of utmost importance in the
existence of cell (whether eukaryotic or prokary-
otic) is the cytoskeleton. The cytoskeleton forms the
scaffold or skeleton at the cellular level in the cyto-
plasm of the cell. The cytoskeleton participates in
a variety of activities of the cell. These involve the
structural matrix of the individual cell and also in
the cellular movement through cilia,1 agella,2 and
lamellipodia.3 The cytoskeleton and its transforma-
tion play vital roles in cell division and intracellular
transportation by facilitating intracellular vesicular
trafc. The fact that the shape of different cell types
is determined by the organization of a network of
tubules (eventually termed cytoskeleton) was estab-
lished by Nikolai Koltsov, in 1903. The term cyto-
skeleton (French: cytosquelette) was rst introduced
in 1931 by the French embryologist Paul Wintrebert.
The association of cytoskeletal proteins with the
plasma membrane and its rearrangement has a very
interesting and well-elaborated role in cell locomo-
tion,4 translocation, and defense mechanisms of the
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
256
cell, such as phagocytosis. The cytoskeleton has a
well-dened role in spindle formation in cell divi-
sion5 and also in pili formation during conjugation
of the prokaryotic cell.6
In the past two decades, researchers have in-
vestigated how the changes in the cell are governed
by the changes and modications in the cytoskel-
eton. The changes signify the overall reprogram-
ming of the cell while differentiating in the course
of tissue- or organ-level compartmentalization.7
The various subtypes of the cell and the cellular
organization, originating from the same germ lay-
er, are largely inuenced by their structural pattern,
which again is governed by cytoskeletal proteins.8
There has been increased emphasis on the role of
cytoskeleton in the development, growth, and dif-
ferentiation of the cell and its various types. The
cytoskeleton and its ultra-structural complexities
constitute not only the gross framework of a cell
but also the localization and functioning of its or-
ganelles.9
There has been a recent re-emergence of em-
phasis on the concepts of neural cytoskeleton. The
study of degeneration in the CNS and the neural
niche throughout different organisms has exposed
many myths about the nervous system. The dogma
that the CNS consists of immutable neurons whose
loss is permanent has been disproved by the ad-
vances in regeneration studies.10 The present evi-
dence clearly shows the presence and replacement
of neurons in the organism throughout life, at dif-
ferent levels. Among the various tissue types in the
body, the neural tissue exhibits the maximum het-
erogeneity,11 and hence the role of cytoskeleton at
both developmental and functional levels is very
important. The polarization of the neuron, con-
sisting of axonal and dendritic development and
arrangement has a series of mechanical and bio-
chemical cues in the rearrangement of the various
cytoskeletal proteins.12 The cytoskeleton plays a
major role in the formation and disruption of the
neural network, and in the formation, viability, and
degeneration of the neurons.
The retina, with its well-dened layers, has
been established as a good model with which to
study the CNS, due to its common ectodermal ori-
gin. The neural retina is extensively studied for its
nature, structure, function, and physiology and has
been manipulated in various ways in studying the
disease models.
This review focuses on the cellular arrange-
ment of neural retina and the pattern of different
cytoskeletal proteins participating in the develop-
ment, disease, and regeneration of the retina.
II. COMPOSITION OF THE CYTOSKELETON
Neurocytoskeleton is the protein skeleton distribut-
ed throughout the cytoplasm that serves such func-
tions such as providing mechanical strength, cell
shape, morphology, locomotion, and transportation
of organelle. Eukaryotic cells contain mainly three
types of cytoskeleton laments (microlament, in-
termediate lament, and microtubule), which show
dynamic behavior by their assembly and disassem-
bly. Microlaments are polymers of actin-forming
bundles and three-dimensional structures. The as-
sembly and disassembly of microlament is regu-
lated by the various actin-binding proteins. Inter-
mediate lament, mainly type-VI nestin, expresses
in dividing neuronal cells during development. Its
expression is higher in neuronal stem cells and di-
minishes in adult neurons. In adulthood, the injury
of CNS enhances expression of nestin.
Microtubule is the third component of cyto-
skeleton that is a polymer of tubulin protein. The
neuronal morphology shows a distinct arrange-
ment of microtubules. Axonal transportation re-
quires the assistance of the microtubule-associated
proteins kinesin and dynein. Kinesin is a coiled
structure with globular head that interacts with
microtubules to move toward the plus end while
dynein moves toward the minus end. Both kinesin
and dynein move opposite each other. Interruption
in axonal transport due to mutation in these mi-
crotubule-associated proteins is the key player in
mediating neuropathological states, as in the cases
of Alzheimer’s and Parkinson’s disease.
On close observation of the micrograph of a
cell, a dense disorganized network of laments
can be seen. There is a marked distinction in the
regions of cell protrusions or in the junctions of
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 257
two adherent cells. These regions have laments
concentrated into bundles. These bundles further
spread out into the interiors of cells as a network
of laments. Bundles and networks form the most
common arrangements of cytoskeletal laments
in a cell. Bundles and networks structurally dif-
fer in the organization of laments. The laments
are packed closely into arrays in bundles, which
are arranged loosely in a crisscross in the case of
networks. Networks further show planar (two-di-
mensional) or net-like arrangements as in nuclear
and plasma membrane, and three-dimensional ar-
rangements, which, for example, gives cytosol its
gel-like properties. In each of these arrangements,
the laments are held together by various cross-
linking proteins.
Actin laments are responsible for the distinct
shape of any cell. The organization of the actin
laments and their associated proteins that con-
nect the microlaments to the membrane govern
the cell morphology. On attachment to a bundle of
laments, the membrane acquires the shape of a
microvillus.
III. RETINA AS A MODEL FOR STUDYING THE
CENTRAL NERVOUS SYSTEM
As an extension of the central nervous system, the
retina, with its well-studied layers, provides a rela-
tively simpler and convenient tool for investigating
the complex central nervous system. Relatively, it
can be easily manipulated from the outside (unlike
brain), making it feasible to test the homing and re-
generative potential of various types of stem cells,
pharmacological compounds, and neurotropic fac-
tors. The common ectodermal origin of retina and
brain during embryonic development makes retina
an excellent experimental tool for studying the
CNS. The similarity in cellular architecture of ret-
ina across various species of experimental organ-
isms has further led to a wide range of organisms
being used for retina-based investigations.
Mammalian retina is composed of seven dis-
tinct layers consisting of different neurons and
glia: a ganglion cell layer, amacrine cells, bipolar
cells and muller glia in inner nuclear layer that act
as inter neurons, rod and cone photoreceptors in
the photoreceptor layer (also considered the outer
nuclear layer), and the retinal pigment epithelial
layer. The axons of ganglion cells traverse through
the optic nerve and connect to the visual cortex of
the brain. These different and highly diverse cell
types arise from the population of retinal progeni-
tor cells, which are a population of quiescent and
actively dividing cells. Recent research has sought
to elucidate the mechanisms controlling the com-
plex processes of neuronal differentiation from the
same source.
IV. NEURONAL/AXONAL DEVELOPMENTAL
CASCADE (FIG. 1)
The cytoskeleton plays a very vital role in neuronal
development. The polarity of the neural cell is es-
tablished by the activity and expression of various
intermediate and associated proteins. Structurally,
neurons have a polarized morphology with two
types of processes, i.e., a long and slender axon and
several short and tapered dendrites. Neurons are not
in the polarized morphology in the course of their
development. They are a result of asymmetrical
cell division of the neural precursors, which is con-
trolled by cytoskeleton.13 The present understand-
ing of the role and contribution of cytoskeleton-
associated proteins in the cellular mechanisms is
not enough. One of the examples that can best sub-
stantiate this fact is the structural proteins like tau,
a microtubule-binding protein that has been exten-
sively studied in the eld of neurodegenerative dis-
eases. Its molecular mechanisms and functions are
well elaborated, but the function within the normal
or diseased neurons has not yet been elucidated.14,15
The study of cytoskeleton development in neurons
can be best reviewed by examining the dynamics
of axonal growth, although the available literature
is relatively scarce. Axons, among all cell types,
are the longest in mature and full-grown animals.
Axons propagate (electrical) messages toward the
synaptic connections in the target cells several
meters away through the neuronal cell body. The
unusual architecture of the axonal shafts is main-
tained by the bundles of parallel microtubules.
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
258
This provides support for structural and long-dis-
tance transport.16 Microtubules in axons are not
continuous and, unlike most cells, are not attached
to the centrosome for their organization.17 The plus
ends of the microtubules mostly point distally.18–20
Short actin laments, organized by spectrin (scaf-
folding protein) into repetitive rings, line the cor-
tex of axon shafts. This linkage to the axonal cell
membrane is again brought about by a second set
of proteins called ankyrins.21 Ankyrins are the an-
chorage and nodal proteins which show robustness
in maintaining the axonal integrity as discussed in
the mouse model of cerebral hypoperfusion.22 The
specic size and shape (varying diameters) of the
different classes of axons are regulated further by a
number of intermediate laments.23 Axons have a
very long span of life, and the dying neurons are a
characteristic feature of the ageing brain.24 Severe
disorganization in the axonal microtubule bundles
are believed to trigger the axonal loss.25,26 The
structural microtubule-binding proteins such as tau
and MAP1B are believed to stabilize the axonal
bundles.27 Moreover, continuous polymerization of
microtubules has been reported in mature axons,
maintaining the steady turnover of microtubules.28
The structural plasticity of neurons demonstrated
by the shortening and lengthening,29–31 the branch-
ing, the re-growth, and the axonal pruning that oc-
curs for sizing the overgrowth,32 are all important
in the regeneration of the damaged neurons. This
regeneration and its associated processes are very
important in synapse formation related to learning
and memory.33,34 The formation of small neuritic
protrusions on the neuronal cell bodies mark the
onset of the formation of axons. The migration of
microtubules by pushing beyond the cell body and
the removal of barrier function is dynamically as-
sisted by the rearrangement of the cortical actin
cytoskeleton35–37 and the formation of lopodia for
extension of microtubules into the membrane.38,39
In a vertebrate neuron, an axon is formed from the
extension of a single neurite out of the many neu-
rites formed on the cell body simultaneously. This
selective process is termed neuron polarization,
and it involves a signaling cascade of cell polar-
ity factors for targeted and specic localization,40
which also requires specicity of stabilization of
microtubules of the selected neurite.41,42
FIG. 1: Hematoxylin & Eosin stain of mouse eye cryosection
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 259
After their establishment, axons follow a ste-
reotype in growth toward their target cell. This
process is generated and executed by growth cones
at the axon tips. The spatio-temporal pattern of
chemical signaling guides the growth cones to-
ward their specic target tissue.43 These guiding
signals have been identied and well elaborated
in their mechanism,44,45 controlling the behavior
of growth cone. The morphogenetic movements,
such as growth velocity, pausing, turning, retrac-
tion or collapse downstream of guidance signal-
ing, are achieved by the particular arrangement of
growth cone actin and microtubule cytoskeleton.46
The growth cones can be divided into three
different zones: the actin-rich peripheral zone; cen-
tral zone, rich in microtubules; and the transition
zone of microtubule and F-actin overlap. The mi-
crotubules spread into the peripheral zone starting
from the center of the growth cones comprising the
axonal tips. The peripheral zone consists of actin-
rich membrane protrusions, which include nger-
like lopodia and veil-like lamellipodia. Filopodia
(containing parallel F-actin bundles) and lamelli-
podia (containing F-actin lattices), after guidance
from the peripheral zone, present their receptors
on their surface. The continuous aggregation and
assembly of actin laments at the leading edge
constantly changes the shape. These laments ow
back in a reverse orientation, depolymerize, and
recycle further in the transition zone. Filopodia
then reach into the growth cone and behave as sig-
naling and adhesion sensors.47–50 The microtubule
behavior in guided axon extension is essentially in-
uenced by this series of events. Lamellipodia acts
as site of substrate adhesion and myosin-II–driven
contraction and force generation.
In another model of growth-cone guidance, the
stabilization of the single microtubule in the pe-
ripheral zone is triggered by external signals,51,52
which are further joined by a bulk of microtubules
from the center, giving rise to a new axonal seg-
ment. The nal step in elongation is the relocation
of the lamellipodia and lopodia in the axon tip.
Microtubules have a crucial role in the implemen-
tation of axon extension in this model, while the
directionality of the extension is achieved by F-
actin. Notably, therefore, the blocking of microtu-
bule dynamics results in the inhibition of axonal
growth.53–55 On the other hand, the inhibition of
actin polymerization either by pharmacological or
by genetic approaches had no suppression of the
axonal growth, in vitro or in vivo. Such interfer-
ence showed some aberration in the turning and
path-nding dynamics of the growth cone.56,57–61
In this regard, it is interesting to note that the
role of F-actin is not always essential in the di-
rectional movement of growth cones. Many sig-
naling mechanisms are targeted to microtubules.
Phosphorylation of some signaling molecules, like
MTBPs APC1, MAP1B, and/or CLASP, mediated
by GSK3 directly regulate the stability of microtu-
bules in growth cones.62–64
The generation of neurons and their differenti-
ation followed thereafter is regulated by a number
of intrinsic and extrinsic factors. The proteins be-
longing to the basic helix–loop–helix (bHLH) fam-
ily like Neurogenin 2 (Ngn2) or Mash (mammalian
achaete-scute complex)-1 or Math (mammalian
atonal homolog)-1 are some of the pro-neural genes
reported in neurogenesis. These mechanisms are
all regulated necessarily by an array of distinct
signaling molecules like Notch signaling, trans-
forming growth factor (TGF)-β/bone morphogenic
protein (BMP) and broblast growth factor (FGF).
The formation and generation of different types of
neuronal types from their progenitors is regulated
by regulatory proteins. The cell cycle is switched
off to regulate number of cells post neurogenesis.65
The regulation of cell cycle in this context is ex-
tremely important for the development of the ner-
vous system. The patterning of neural tissue is de-
termined by SRY (sex-determining region Y) box,
Sox, and the Pax (paired box) family of proteins.
Pax6 (a paired-box transcription factor), which is
highly conserved among vertebrate development,
is responsible for the overall patterning of the em-
bryonic nervous system.66 It directly regulates the
expression of Ngn2 and also upregulates the ex-
pression of cell adhesion molecules as well as the
cell-cycle inhibitors such as p27.67 This eventually
links together the differentiation and the withdraw-
al from the cell cycle. Again, the maintenance of
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
260
neural cells in the undifferentiated state is brought
about by Sox1, Sox2, and Sox3 proteins via sup-
pression of neurogenesis.68
Coordination of transcriptional program in the
neurogenesis occurs also at the epigenetic level.
The overexpression of HDAC4 (histone deacety-
lase) in granule neurons, shields them from apop-
tosis without undergoing the PI3K or ERK/MAPK
pathways. The phenomenon of neuroprotection is
brought about by blocking the abnormal progres-
sion of cell cycle by the inhibition of CDK1 (cell
cycle kinase).69
The overall transcriptional programs and epi-
genetic regulation is mediated by signal transduc-
tion pathways. The signal transduction pathways
are further regulated by the events in cell cycle.
The essential role of Notch signaling in neurogen-
esis has been demonstrated both in vitro70 and in
vivo.71 It inhibits the neural gene expression later-
ally, and hence limits the number of differentiating
neurons. In vertebrate neurogenesis, the notch and
delta signaling are restricted only to some stages in
the events of cell cycle.72
V. THE GROWTH AND DIFFERENTIATION
DYNAMICS
After exit from the cell, the generation of intri-
cate networks of the microtubule in neurons and
the extensions has to be brought about by a well-
orchestrated differentiation program. This is also
brought about by the timed entry and exit of the
cell cycle. While studying the mutations in dynac-
tin in a zebrash model, a premature cell cycle exit
and acceleration of neurogenesis was exhibited by
the retinal progenitors. The cell signaling also gov-
erns cell fate. The defects in trafcking of Notch
to the apical region of retinal progenitors has been
reported in affecting the early cell fates at the ex-
pense of late ones.73
Neurons migrate to their functional location
from the site of their origin via polarized move-
ment of the cytoskeleton.74 In the case of neuro-
genesis in the retina, a specic pattern is followed.
During the development of the retina, retinal gan-
glion cells are the rst to differentiate, then cones,
followed by horizontal cells. After these, the rod
cells and the amacrine cells are formed. The last
cells to differentiate are the bipolar cells. Retinal
neurons in mammals differentiate before birth.75–77
Neurogenesis of retinal ganglion cells is deter-
mined intrinsically. A number of genes have been
identied in this process of neurogenesis.78 The
deletion of Pax6, Barhl2, or Math5 reduces and/
or blocks the differentiating RGCs in mice.79, 81–83
VI. CYTOSKELETAL PROTEINS IN THE RETINA
AT DIFFERENT LEVELS
From the beginning, understanding the cell-fate
specications and mechanisms has remained a
challenge in the study of retinal development. The
Rho family plays a key role in cytoskeletal arrange-
ments in the RPE in the TGF-β1–induced pathway.
It has been demonstrated experimentally that treat-
ment with TGF-β1 induces cytoskeleton reorga-
nization, expression of α-SMA, an increase in the
phosphorylation of ERK, Smad, AKT, and activa-
tion of Rac1 and RhoA. These mechanisms are in-
hibited by treatment with Rho inhibitors. TGF-β1
also increases LIMK and colin phosphorylation.
Based on these observations, it has been well pro-
posed and established that the cytoskeletal actin re-
arrangement in human RPE is induced by TGF-β1
via the Rho GTPase-dependent pathways modulat-
ing LIM kinase and colin activity.84
The role of cytoskeleton proteins can also be
well elaborated in the phagocytosis mechanism of
the RPE. The shed photoreceptor outer segments
are phagocytosed by the cells of the RPE (retinal
pigment epithelium) through a mechanism that
involves αv integrins upstream of Rho GTPases
and tyrosine kinases. Retinal pigment epithelial
cells have the tendency to activate and redistribute
Rac1, but they do not do the same with RhoA or
Cdc42 during phagocytosis. The particle engulf-
ment, in this case by the RPE, is prevented when
dominant-negative Rac1 is overexpressed as well
as when Rac1 expression is decreased. It has been
further experimentally demonstrated that αvβ5
integrin and its ligand MFG-E8 (milk fat globule
EGF factor-8), are required for the activation of
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 261
Rac1 during phagocytosis, but they do not involve
the receptor tyrosine kinase MerTK. FAK inhibi-
tion hinders the tyrosine kinase signaling down-
stream of αvβ5 toward MerTK. This specic hin-
drance does not prevent Rac1 activation nor does
it prevent the recruitment of F-actin during phago-
cytosis. Inhibition of Rac1 does not effect FAK or
MerTK activation. Therefore, activation/recruit-
ment of MerTK and F-actin via FAK and Rac1,
respectively, require MFG-E8–ligated αvβ5 integ-
rin. These pathways have independent activation
and are essential for clearance phagocytosis. The
major source of endothelin-1 (ET-1) at the blood
retinal barrier is the retinal pigment epithelium
(RPE). Thrombin can act on the RPE and can prob-
ably injure the blood retinal barrier. The activation
of protein kinase C and the transient intracellular
calcium mobilization brought about by thrombin
probably play a minor role in endothelin-1 synthe-
sis in the retinal pigment epithelium.85
The outgrowth of the neurites, and eventually
the growth cone repulsion, are mediated by a mem-
ber of the subfamily of cell adhesion molecules,
i.e., protein tyrosine phosphatase (RPTP). PTPμ
acts as a permissive substrate for the growth of
retinal ganglion cell (RGC) neurites while it inhib-
its the growth of temporal retinal ganglion cells.
In temporal neurons, Rac 1 acts as the regulating
switch between the permissive and repulsive re-
sponses to PTPμ. For the repulsion of temporal
neurons, Rac 1 needs to be inhibited. Being an
inhibitor for nasal growth, the activity of Cdc42
is essential for both permissive and repulsive re-
sponses to PTPμ. However, inhibition of Rac1 ac-
tivity is the key to PTPμ-dependent repulsion in
temporal RGC neurons.86
Studies on the activity of actin/colin proteins
have shown an active role in axonal extension87–91
and have an implications in the regulation of neu-
rotrophins on lopodial responses.92,93 A recent
study showed that the direct role of actin/colin
proteins in response to bone morphogenic proteins
displays growth-cone guidance in the spinal neu-
rons of Xenopus laevis.94 Another very important
member of this group of molecules is the Rho fam-
ily of GTPases. Of the various molecules amongst
the small GTPase related to Rho, Rac1 is involved
in the regulation of cytoskeletal dynamics and is
critically involved in the development of neuron,
axonal growth, and cell survival. Rac1 in its con-
stitutively active form has been shown to promote
the growth of axons in inhibitory environments of
in vivo95 as well as in vitro96 models. It has been
reported to promote survival in vivo as well.97, 98 In
studies involving the developing chick retina, NF-
M, i.e., mid-size neurolament99–101 and a chick-
specic MAP (microtubule associated protein),
have been extensively studied to identify ganglion
cells and ganglion precursor cells.102 It has been
demonstrated that the expression of MAP(RA4)
and NF-M proteins in the chick retina is not re-
stricted to ganglion cells alone as otherwise thought
and accepted. Rather, it is established that these
proteins are expressed transiently in a develop-
mentally regulated manner by all neuronal retinal
progenitors.103 The Lim-kinase (LIMK) proteins
essentially regulate the actin cytoskeleton. It is
therefore believed that they control many process-
es, including cell cycling, cell integrity, migration
and axonal guidance, during development.104 The
loss of LIMK1 resulted in defects in the neuronal
growth cone, dendritic spines, and the organiza-
tion of actin. These results were demonstrated by
the different experiments in-vitro and in-vivo and
resulted in abnormal long-term potentiation of the
hippocampus and synaptic changes.105 Glycogen
synthase kinase (GSK3) regulates a large number
of transcription factors, including β-catenin, c-Jun,
and CREB (cAMP response element-binding pro-
tein), to name a few.106 A study involving the regu-
lation of transcription factors by GSK3β shows the
involvement of kinase essentially in the pruning of
developmental axon both in vitro and in vivo.107 A
wide range of cytoskeleton dynamics and several
types of purinergic receptors are controlled by ex-
tracellular ATP. An intracellular Ca(2+) increase,
induced by the ATP, follows colin rod formation.
Cells that express another variant of colin (un-
phosphorylatable) do not exhibit ATP-induced co-
lin rod formation. The formation of colin rods is
induced by the calcineurin-dependent dephosphor-
ylation of colin induced by the inux of Ca(2+)
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
262
through P2X receptors. This pathway may essen-
tially elucidate how ATP effects development, de-
generation, and injury of a neuron.108
VII. ROLE OF CYTOSKELETON IN NEURAL/
NEURONAL DEVELOPMENT IN ANIMAL
MODELS
The study of neuronal polarity and synaptogenesis
and associated proteins has been studied in Dro-
sophila, Caenorhabditis elegans, Xenopus, and
zebrash models. A study of Drosophila demon-
strated that when Cyp, a cytoplasmic FMRP-
interacting protein, is mutated, it inhibits the l-
amentous actin (F-actin) to assemble, which is a
key regulating aspect of synaptogenesis. The study
also proposed that Cyp inhibits actin assembly
as well, regulating the development of synapse as
well as endocytosis.109 Intraagellar transport (IFT)
is essential in developing the ciliated sensory outer
segments (OS), which sequester a number of pro-
teins in the vertebrate phototransduction machin-
ery. Rho GTPases are essential regulators of cyto-
skeletal reorganization in the case of Drosophila as
well. Colin is a protein responsible for the depo-
lymerization of actin, very essential in the process
of neurogenesis. It is regulated by the inhibition
of LIM kinase and activation of Slingshot phos-
phatase. During Axon growth, the genetic analysis
of the different pathways have revealed conver-
gent and divergent pathways from Rho GTPases
to the cytoskeleton achieved by biases in pathway
selection.110 In one of the experiments involving
Drosophila, neuronal cells in vitro demonstrated
that the regulation of cellular actin dynamics, the
deciency of pantothenatekinase (PANK), a cata-
lyst that converts pantothenic acid to co-enzyme
A, leads to abnormal organization in the F-actin. A
reduction in in the activity of PANK up-regulates
the phosphorylated form of colin, which is a con-
served protein responsible for the severing of actin
lament.111
However, the mechanism of the cytoskeletal
proteins and the associated proteins/molecules have
been studied in the C. elegans model. In a study
using C. elegans, it was demonstrated that during
axonal outgrowth, the Arp2/3 complex regulates
the initiation of growth cone lopodia.112 The axon
guidance in C. elegans has been shown to be af-
fected by UNC-115/abLIM and UNC-34/Enabled,
which are actin modulatory proteins.113–115 Enabled
essentially regulates the formation of lopodia in
neurons. It probably acts by blocking the actin-
capping activity, resulting in long lament growth
in lopodia.116,117 Some other studies show that En-
abled might express the anti-capping independent
roles118 involved in lopodia formation.11 9 OSM-3,
belonging to the family of homodimeric kinesin 2
motor, has an essential role in a few sensory cil-
ia.120,121 Kif17, a homologue for the OSM-3 gene in
vertebrates, is known to be responsible for the traf-
cking of dendrites in neurons. Its role in the pro-
cess of ciliogenesis has not yet been dened. Kif17
is widely expressed in the zebrash nervous sys-
tem and retina.122 Sufcient evidence has proven
the role of ADF (actin depolymerizing factor) and
colin in the regulation of axonal growth and actin
dynamics. The expression of ADF/colins is high
in neuronal growth cones, especially in the areas
where there is a high rate of actin disassembly.123
Furthermore, the overexpression of these proteins
in a Xenopus model showed a nearly 50% increase
in the length of neurites.124
VIII. SNPS IN CYTOSKELETON IMPAIRMENT
Various molecules have been studied to acquire
deeper mechanistic understanding of retinal de-
generation. The mutations and/or deletions or
single nucleotide polymorphisms seen in the mol-
ecules during disease or degeneration lack proper
co-relation with the cytoskeleton. The dynamics of
the cytoskeleton plays a critical role in neural-crest
cell migration. One important molecules discussed
from the beginning of this review is colin. The
dynamics of cytoskeleton components, especially
in neural tube development, is regulated by colin.
Colin is encoded by the gene C1, and its poly-
morphisms have been investigated and established
as a risk factor of spina bida. In a population-based
control study in California of non-Hispanic white
infants with spina bida, ve SNPs in the gene
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 263
enhanced disease risk by greater than two-fold.125
The regulation of the cytoskeleton is essentially
brought about by protein–protein interactions.
The interaction of integrin and actin is regulated
by the FLNA gene. Mutations in the FLNA gene
result in congenital malformations in craniofa-
cial structures.126 In one genetic study of two sib-
lings having an unusual set of disease conditions
(i.e. optic atrophy, early-onset spastic paraplegia,
and neuropathy), one homozygous c.316C>T(p.
R106C) variant in the Trk-fused gene (TFG) was
the only plausible mutation upon linkage analysis,
genome-wide SNP-typing, and exome sequencing.
The characterization of this protein did not yield
any self-assembly into an oligomeric complex,
which is essential for the normal functioning of
TFG. The inhibition of TFG alters endoplasmic
reticulum (ER) morphology while slowing down
its protein secretion. This results in the disruption
of peripheral tubules and eventually the ER col-
lapses over the cytoskeleton beneath it. This study
highlights the link between the involvement of
cytoskeleton through altered ER architecture and
neurodegeneration.127 TUBA8 gene, encodes a
variant of α-tubulin with unknown function and
it regulates the function of microtubules during
the migration of cortical neurons. One study has
reported that the mutation of this gene results in
generalized polymicrogyria along with optic nerve
hypoplasia (PMGOH). This study suggests the role
of TUBA8 in the regulation of brain development
in mammals.128 Mutations in PFN1, a gene encod-
ing the actin monomer-binding protein prolin 1,
were recently reported in 1–2% of familial amyo-
trophic lateral sclerosis (ALS) patients. Two rare
non-synonymous variants (E117D and E117G)
have been identied in sporadic ALS patients.129
Cerebral PGV (proliferative glomeruloid vascu-
lopathy) is a severe autosomal recessive disorder
of brain angiogenesis that results in abnormal
thickening and unusually perforated vessels which
lead to hydranencephaly. Mutation and deletion in
the FLVCR2 gene, which encodes a 12-transmem-
berane domain constituting a putative transporter,
results in the absence of alpha-smooth muscle ac-
tin, suggesting a decit in pericytes.130
IX. CONCLUSION
The eld of retinal neural cytoskeletal dynamics
in the growth and development of the vertebrate
retina needs denition. This review highlights the
role of retinal architecture and the involvement of
cytoskeletal proteins in the growth and differentia-
tion of the neuronal cells, thus indicating the need
for translational research. Therefore, the study of
retinal development, growth, and differentiation is
likely to contribute signicantly in the context of
functional retinal repair. The mechanisms underly-
ing differentiation and compartmentalization pro-
vide new insights into retinal development. In this
regard, regenerative approaches in stem cell trans-
plantation need to integrate such methodologies to
account for poor results and irreproducible data.
REFERENCES
1. Burgoyne T, Dixon M, Luther P, Hogg C, Shoemark A.
Generation of a three-dimensional ultrastructural model
of human respiratory cilia. Am J Respir Cell Mol Biol.
2012;47:800–6.
2. Lin J, Heuser T, Song K, Fu X,Nicastro D. One of the
nine doublet microtubules of eukaryotic agella exhibits
unique and partially conserved structures. PLoS One.
2012;10: e46494.
3. Ingerman E, Hsiao JY, Mullins RD. Arp2/3 complex ATP
hydrolysis promotes lamellipodial actin network disas-
sembly but is dispensable for assembly. 2013;5:619–33.
4. Samoilov VO, Bigdai EV, Kryzhanovskii EV, Dudich
BA, Panov SA, Smirnov DD, Nesterov AS. Respiratory
and olfactory cilia molecular machines of locomotor ap-
paratus. Biozika. 2013;2:269–75.
5. Wang H, Brust-MascherI, Cheerambathur D, Scholey
JM. Coupling between microtubule sliding, plus-end
growth and spindle length revealed by kinesin-8 deple-
tion. Cytoskeleton. 2010; 11:715–28.
6. Cowles KN, Gitai Z. Surface association and the MreB
cytoskeleton regulate pilus production, localization and
function in Pseudomonas aeruginosa. Mol Microbiol.
2010;6:1411–26.
7. Chen J, Lippincott-Schwartz J, Liu J. Intracellular spatial
localization regulated by the microtubule network. PLoS
One. 2012; 4:e34919.
8. Krainer EC, Ouderkirk JL, Miller EW, Miller MR,
Mersich AT, Blystone SD. The multiplicity of human
formins: Expression patterns in cells and tissues. Cyto-
skeleton. 2013.
9. Tervaniemi MH, Siitonen HA, Söderhäll C, Minhas G,
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
264
Vuola J, Tiala I, Sormunen R, Samuelsson L, Suomela S,
Kere J, Elomaa O. Centrosomal localization of the pso-
riasis candidate gene product, CCHCR1, supports a role
in cytoskeletal organization. PLoS One 2012;11:49920.
10. Ma WP, Li YT, Tao HW. Down regulation of cortical in-
hibition mediates ocular dominance plasticity during the
critical period. J Neurosci. 2013;27:11276–80.
11. Liu X, Zhang Z, Ribelayga CP. Heterogeneous expres-
sion of the core circadian clock proteins among neuronal
cell types in mouse retina. PloS One. 2012;11:50602.
12. Fukata M, Nakagawa M, Kaibuchi K. Roles of Rho-fam-
ily GTPases in cell polarisation and directional migra-
tion. Curr Opin Cell Biol. 2003;5:590–7.
13. Knoblich JA. Mechanisms of asymmetric stem cell divi-
sion. Cell. 2008;132:583–97.
14. Hirokawa N, Niwa S, Tanaka Y. Molecular motors in neu-
rons: transport mechanisms and roles in brain function,
development, and disease. Neuron. 2010;68:610–38.
15. Morris M, Maeda S, Vossel K, Mucke L. The many faces
of tau. Neuron. 2011;70:410–26.
16. Twelvetrees A, Hendricks AG, Holzbaur ELF. SnapShot:
axonal transport. Cell. 2012;149:950.
17. Kuijpers M, Hoogenraad CC.Centrosomes, microtu-
bules and neuronal development. Mol Cell Neurosci.
2011;4:349–58.
18. Ahmad FJ, Yu W, McNally FJ, Baas PW. An essential
role for katanin in severing microtubules in the neuron. J.
Cell Biol. 1999;145:305–15.
19. Baas PW, Lin S. Hooks and comets: The story of micro-
tubule polarity orientation in the neuron. Dev. Neurobiol.
2011;71:403–18.
20. Stiess M, Maghelli N, Kapitein LC, Gomis-Rüth
S, Wilsch-Bräuninger M, Hoogenraad C C,Tolić-
Nørrelykke IM, Bradke F. Axon extension occurs inde-
pendently of centrosomal microtubule nucleation. Sci-
ence. 2010;327:704–7.
21. Xu K, Zhong G, Zhuang X. Actin, Spectrin, and asso-
ciated proteins form a periodic cytoskeletal structure in
axons. Science 2013; Reimer MM, McQueen J, Searcy
L, Scullion G, Zonta B, Desmazieres A, Holland PR,
Smith J, Gliddon C, Wood ER, Herzyk P, Brophy PJ, Mc-
Culloch J, Horsburgh K. Rapid disruption of axon-glial
integrity in response to mild cerebral hypoperfusion. J
Neurosci. 2011;49:18185–94.
22. Perrot R, Eyer J. Neuronal intermediate laments
and neurodegenerative disorders. Brain Res Bull.
2009;80:282–95.
23. Marner L, Nyengaard JR, Tang Y, Pakkenberg B. Marked
loss of myelinated nerve bers in the human brain with
age. J Comp Neurol. 2003;462:144–52.
24. Adalbert R, Nogradi A, Babetto E, Janeckova L, Walk-
er SA, Kerschensteiner M, Misgeld T, Coleman MP.
Severely dystrophic axons at amyloid plaques remain
continuous and connected to viable cell bodies. Brain.
2009;132: 402–16.
25. Fiala JC, Feinberg M, Peters A, Barbas H. Mitochondrial
degeneration in dystrophic neurites of senile plaques
may lead to extracellular deposition of ne laments.
Brain Struct. Funct. 2007; 212:195–207.
26. Chilton J, Gordon-Weeks P. Role of microtubules and
MAPs during neuritogenesis. In Intracellular Mecha-
nisms for Neuritogenesis de Curtis, I Springer. 2007;
ed57–88.
27. Kollins KM, Bell RL, Butts M, Withers GS. Dendrites
differ from axons in patterns of microtubule stability
and polymerization during development. Neural Dev.
2009;4:26.
28. Küppers MB, Letzkus JJ, Lüer K, Technau G, Schmidt
H, Prokop A. A new culturing strategy optimises Dro-
sophila primary cell cultures for structural and functional
analyses. Dev. Biol. 2004;269:459–78.
29. Lamoureux P, Heidemann SR, Martzke NR, Miller KE.
Growth and elongation within and along the axon. Dev.
Neurobiol. 2010;70:135–49.
30. Rajagopalan J, Tofangchi A, Saif MT. Drosophila neu-
rons actively regulate axonal tension in vivo. Biophys. J.
2010;99:3208–15.
31. Letourneau PC. Actin in axons: stable scaffolds and dy-
namic laments. Results Probl.Cell Differ. 2009;48:265–
90.
32. Saxena S, Caroni P. Mechanisms of axon degenera-
tion: from development to disease. Prog. Neurobiol.
2007;83:174–91.
33. Bradke F, Fawcett JW, Spira ME. Assembly of a new
growth cone after axotomy: the precursor to axon regen-
eration. Nat. Rev. Neurosci. 2012;13:183–93.
34. Flynn KC, Hellal F, Neukirchen D, Jacob S, Tahirovic
S, Dupraz S, Stern S, Garvalov BK, Gurniak C, Shaw
AE, Meyn L, Wedlich-Söldner R, Bamburg JR, Small
JV, Witke W, Bradke F. ADF/colin-mediated actin ret-
rograde ow directs neurite formation in the developing
brain. Neuron 2012;76:1091–107.
35. Neukirchen D, Bradke F. Neuronal polarization and the
cytoskeleton. Semin. Cell Dev. Biol. 2011;22:825–33.
36. Saengsawang W, Mitok K, Viesselmann C, Pietila
L,Lumbard DC, Corey SJ, Dent EW. The F-BAR protein
CIP4 inhibits neurite formation by producing lamellipo-
dial protrusions. Curr. Biol. 2012;22: 494–501.
37. Dent EW, Kwiatkowski AV, Mebane LM, Philippar U,
Barzik M, Rubinson DA, Gupton S, Van Veen JE, Fur-
man C, Zhang J, Alberts AS, Mori S, Gertler FB. Filopo-
dia are required for cortical neurite initiation. Nat. Cell
Biol. 2007;9:1347–59.
38. Gonçalves-Pimentel C, Gombos R, MihályJ, Sánchez-
Soriano N, Prokop A. Dissecting regulatory networks of
lopodia formation in a Drosophila growth cone model.
PLoS One. 2011;6:e18340.
39. Barnes AP, Polleu F. Establishment of axon-dendrite
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 265
polarity in developing neurons. Annu. Rev. Neurosci.
2009;32:347–81.
40. Brandt R. Cytoskeletal mechanisms of axon outgrowth
and pathnding. Cell Tissue Res. 1998;292:181–9.
41. Chen S, Chen J, Shi H, Wei M, Castaneda-Castellanos
DR, Bultje RS, Pei X, Kriegstein AR, Zhang M, Shi SH.
Regulation of microtubule stability and organization by
mammalian Par3 in specifying neuronal polarity. Dev.
Cell. 2013;24:26–40.
42. Tessier-Lavigne M, Goodman CS. The molecular biol-
ogy of axon guidance. Science. 1996;274:1123–33.
43. Araújo SJ, Tear G. Axon guidance mechanisms and mol-
ecules: lessons from invertebrates. Nat Rev Neurosci.
2003;4:910–22.
44. Huber AB, Kolodkin AL, Ginty DD, Cloutier JF. Signal-
ing at the growth cone: ligand-receptor complexes and
the control of axon growth and guidance. Annu Rev
Neurosci. 2003;26:509–63.
45. Fan J, Manseld SG, Redmond T, Gordon-Weeks PR,
Raper JA. The organization of F-actin and microtubules
in growth cones exposed to a brain-derived collapsing
factor. J Cell Biol. 1993;121:867–78.
46. Chien CB, Rosenthal DE, Harris WA, Holt CE. Naviga-
tional errors made by growth cones without lopodia in
the embryonic Xenopus brain. Neuron. 1993;11:237–51.
47. Davenport W, Dou P, Rehder V, Kater SB. A sen-
sory role for neuronal growth cone lopodia. Nature.
1993;361:721–24.
48. Dwivedy A, Gertler FB, Miller J, Holt C E, Lebrand
C.Ena/VASP function in retinal axons is required for ter-
minal arborization but not pathway navigation. Develop-
ment. 2007;134:2137–46.
49. Mattila PK, Lappalainen P. Filopodia: molecular archi-
tecture and cellular functions. Nat. Rev Mol Cell Biol.
2008; 9:446–54.
50. Dent EW, Gupton SL, Gertler FB. The growth cone cy-
toskeleton in axon outgrowth and guidance. Cold Spring
Harb Perspect Biol. 2011;3:3.
51. Lowery LA, Van VD. The trip of the tip: understand-
ing the growth cone machinery. Nat. Rev Mol Cell Biol.
2009;10:332–343.
52. Letourneau PC, Ressler AH. Inhibition of neurite ini-
tiation and growth by taxol. J Cell Biol. 1984;98:1355–
1362.
53. Sánchez SN, Gonçalves PC, Beaven R, Haessler U, Ofner
ZL, Ballestrem C, Prokop A. Drosophila growth cones: a
genetically tractable platform for the analysis of axonal
growth dynamics. Dev. Neurobiol. 2010;70:58–71.
54. Tanaka E, Ho T, Kirschner MW. The role of microtubule
dynamics in growth cone motility and axonal growth. J
Cell Biol. 1995;128:139–55.
55. Buck KB, Zheng JQ. Growth cone turning induced by di-
rect local modication of microtubule dynamics. J Neu-
rosci. 2002;22:9358–67.
56. Challacombe JF, Snow DM, Letourneau PC. Actin la-
ment bundles are required for microtubule reorientation
during growth cone turning to avoid an inhibitory guid-
ance cue. J Cell Sci. 1996;109:2031–40.
57. Chien CB, Rosenthal DE, Harris WA, Holt CE. Naviga-
tional errors made by growth cones without lopodia in
the embryonic Xenopus brain. Neuron. 1993;11:237–51.
58. Kaufmann N, Wills ZP, Van VD. Drosophila Rac1 con-
trols motor axon guidance. Development. 1998;125:453–
61.
59. Marsh L, Letourneau PC. Growth of neurites without -
lopodial or lamellipodial activity in the presence of cyto-
chalasin B. J Cell Biol. 1984;99:2041–7.
60. Hur EM, Saijilafu Lee BD, Kim SJ, Xu WL, Zhou
FQ. GSK3 controls axon growth via CLASP-mediated
regulation of growth cone microtubules. Genes Dev.
2011;25:1968–81.
61. Lucas FR, Goold RG, Gordon-Weeks PR, Salinas PC.
Inhibition of GSK-3beta leading to the loss of phos-
phorylated MAP-1B is an early event in axonal re-
modelling induced by WNT-7a or lithium. J Cell Sci.
1998;111:1351–61.
62. Purro SA, Ciani L, Hoyos FM, Stamatakou E, Siomou E,
Salinas PC. Wnt regulates axon behavior through chang-
es in microtubule growth directionality: a new role for
adenomatous polyposis coli. J Neurosci. 2008;28:8644–
54.
63. Maurange C, Cheng L, Gould AP. Temporal transcription
factors and their targets schedule the end of neural prolif-
eration in Drosophila. Cell. 2008;133:891–902.
64. Osumi N, Shinohara H, Numayama-Tsuruta K, Maekawa
M. Pax6 transcription factor contributes to both embry-
onic and adult neurogenesis as a multifunctional regula-
tor. Stem Cells 2008;26:1663–72.
65. Sandberg M, Källström M, Muhr J. Sox21 promotes the
progression of vertebrate neurogenesis. Nat Neurosci.
2005;8:995–1001.
66. Majdzadeh N, Wang L, Morrison BE, Bassel-Duby R,
Olson EN, D’Mello SR. HDAC4 inhibits cell-cycle pro-
gression and protects neurons from cell death. Dev Neu-
robiol. 2008;68:1076–92.
67. Abburi C, Prabhakar S, Kalra J, Huria A, Anand A. Vas-
cular endothelial growth factor (VEGF) induced prolif-
eration of human fetal derived ciliary epithelium stem
cells is mediated by jagged-N cadherin pathway. Curr
Neurovasc Res. 2013;2:93–102.
68. Hu QD, Ma QH, Gennarini G, Xiao ZC. Cross-talk be-
tween F3/contactin and Notch at axoglial interface: a
role in oligodendrocyte development. Dev Neurosci.
2006;28:25–33.
69. Cisneros E, Latasa MJ, García-Flores M, Frade
JM.Instability of Notch1 and Delta1 mRNAs and re-
duced Notch activity in vertebrate neuroepithelial cells
undergoing S-phase. Mol Cell Neurosci. 2008;37:820–
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
266
31.
70. Götz M, Huttner WB. The cell biology of neurogenesis.
Nat Rev Mol Cell Biol. 2005;6:777–88.
71. Marin O, Valiente M, Ge X, Tsai LH. Guiding neuro-
nal cell migrations. Cold Spring Harb Perspect Biol.
2010;2:a001834.
72. Altshuler D, Turner D, Cepko C. Specication of cell
types in the vertebrate retina, Development of the Vi-
sual System (Lam D., Shatz C., eds),1991;pp 37–58 MIT
Press Boston
73. Cepko CL, Austin C P, Yang X, Alexiades M, Ezzedine
D. Cell fate determination in the vertebrate retina. Proc
Natl Acad Sci. 1996;93:589–95.
74. Marquardt T and Gruss P. Generating neuronal diver-
sity in the retina: one for nearly all. Trends Neurosci.
2002;25:32–8.
75. Mu X, Klein WH. Gene regulatory networks and retinal
ganglion cell development. Eye, Retina, and Visual Sys-
tem of the Mouse. PNAS. 2008;19:6942–7.
76. Brown NL, Patel S, Brzezinski J, Glaser T. Math5 is re-
quired for retinal ganglion cell and optic nerve forma-
tion. Development. 2001;128:2497–508.
77. Marquardt T, Ashery PR, Andrejewski N, Scardigli R,
Guillemot F, Gruss P. Pax6 is required for the multipotent
state of retinal progenitor cells. Cell. 2001;105:43–55.
78. Wang SW, Kim BS, Ding K, Wang H, Sun D, John-
son RL, Klein WH, Gan L. Requirement for math5 in
the development of retinal ganglion cells. Genes Dev.
2001;15:24–9.
79. Lee J, Ko M, Joo CK. Rho plays a key role in TGF-beta1-
induced cytoskeletal rearrangement in human retinal pig-
ment epithelium. J Cell Physiol. 2008;216:520–6.
80. Narayan S, Prasanna G, Tchedre K, Krishnamoorthy R,
Yorio T. Thrombin-induced endothelin-1 synthesis and
secretion in retinalpigment epithelial cells is rho kinase
dependent. J Ocul Pharmacol Ther. 2010;26:389–97.
81. Major DL, Brady-Kalnay SM.Rho GTPases regulate
PTPmu-mediated nasal neurite outgrowth and temporal
repulsion of retinal ganglion cell neurons. Mol Cell Neu-
rosci2007;34:453–67.
82. Bamburg JR, Bray DJ. Distribution and cellular lo-
calization of actin depolymerizing factor. Cell Biol.
1987;105:2817–25.
83. Birkenfeld J, Betz H, Roth D. Inhibition of neurite ex-
tension by overexpression of individual domains of LIM
kinase 1. J Neurochem. 2001;78:924–7.
84. Kuhn TB, Meberg PJ, Brown MD, Bernstein BW, Min-
amide LS, Jensen JR, Okada K, Soda EA, Bamburg JR.
Regulating actin dynamics in neuronal growth cones
by ADF/colin and rho family GTPases. J Neurobiol.
2000;44:126–44.
85. Meberg PJ, Bamburg JR. Increase in neurite outgrowth
mediated by overexpression of actin depolymerizing fac-
tor. J Neurosci. 2000;20:2459–69.
86. Meberg PJ, Ono S, Minamide LS, Takahashi M, Bamburg
JR. Actin depolymerizing factor and colin phosphoryla-
tion dynamics: response to signals that regulate neurite
extension. Cell Motil Cytoskeleton. 1998;39:172–90.
87. Chen TJ, Gehler S, Shaw AE, Bamburg, JR, Letourneau
PC. Cdc42 participates in the regulation of ADF/colin
and retinal growth cone lopodia by brain derived neuro-
trophic factor. J Neurobiol. 2006;66:103–14.
88. Gehler S, Shaw AE, Sarmiere PD, Bamburg JR, Letour-
neau PC. Brain-derived neurotrophic factor regulation
of retinal growth cone lopodial dynamics is mediated
through actin depolymerizing factor/colin. J Neurosci.
2004;24:10741–9.
89. Wen Z, Han L, Bamburg JR, Shim S, Ming GL, Zheng
JQ. BMP gradients steer nerve growth cones by a balanc-
ing act of LIM kinase and slingshot phosphatase on DF/
colin. J Cell Biol. 2007;178:107–19.
90. Jain A, McKeon RJ, Brady-Kalnay SM, Bellamkonda
RV. Sustained delivery of activated Rho GTPases and
BDNF promotes axon growth in CSPG-rich regions fol-
lowing spinal cord injury. PLoS One. 2011;6:e16135.
91. Jain A, Brady-Kalnay SM, Bellamkonda RV. Modulation
of Rho GTPase activity alleviates chondroitin sulfate
proteoglycan-dependent inhibition of neurite extension.
J Neurosci Res. 2004;77:299–307.
92. Linseman DA, Laessig T, Meintzer MK, McClure M,
Barth H, Aktories K, Heidenreich KA. An essential role
for Rac/Cdc42 GTPases in cerebellar granule neuron sur-
vival. J Biol Chem. 2001;276:39123–31.
93. Le SS, Loucks FA, Udo H, Richardson-Burns S, Phelps
RA, Bouchard RJ, Barth H, Aktories K, Tyler KL, Kan-
del ER, Heidenreich KA, Linseman DA. Inhibition of
RacGTPase triggers a c-Jun- and Bim-dependent mito-
chondrial apoptotic cascade in cerebellar granule neu-
rons. J Neurochem. 2005;94:1025–39.
94. Bennett GS, DiLullo C. Expression of a neurolament
protein by the precursors of a subpopulation of ventral
spinal cord neurons. Dev Biol. 1985;1:94–106.
95. Grant P, Pant HC. Neurolament protein synthesis and
phosphorylation. J Neurocytol. 2000;29:843–72.
96. Grant P, Sharma P, Pant HC. Cyclin-dependent protein
kinase 5 (Cdk5) and the regulation of neurolament me-
tabolism. Eur J Biochem. 2001;6:1534–46.
97. Poulain FE, Sobel A. The microtubule network and neu-
ronal morphogenesis: dynamic and coordinated orches-
tration through multiple players. Mol Cell Neurosci.
2010;1:15–32.
98. Gutierrez C, McNally M, Canto-Soler MV. Cytoskeleton
proteins previously considered exclusive to ganglion
cells are transiently expressed by all retinal neuronal pre-
cursors. BMC Developmental Biology. 2011;11:46.
99. Lindström NO, Neves C, McIntosh R, Miedzybrodzka Z,
Vargesson N, Collinson JM. Tissue specic characterisa-
tion of Lim-kinase 1 expression during mouse embryo-
Volume 24, Number 3, 2014
Cytoskeleton Dynamics in the Retina 267
genesis. Gene Expr Patterns. 2011;11:221–32.
100. Meng Y, Zhang Y, Tregoubov V, Janus C, Cruz L, Jack-
son M, Lu WY, MacDonald JF, Wang JY, Falls DL, Jia
Z. Abnormal spine morphology and enhanced LTP in
LIMK-1 knockout mice. Neuron 2002;35:121–33.
101. Hur EM, Zhou FQ. GSK3 signalling in neural develop-
ment. Nat Rev Neurosci. 2010;11:539–51.
102. Chen M, Maloney JA, Kallop DY, Atwal JK, Tam S J,
Baer K, Kissel H, Kaminker JS, Lewcock JW, Weimer
RM, Watts RJ. Spatially coordinated kinase signal-
ing regulates local axon degeneration. J Neurosci.
2012;39:13439–53.
103. Homma S, Shimada T, Hikake T, Yaginuma H. Expres-
sion pattern of LRR and Ig domain-containing protein
(LRRIG protein) in the early mouse embryo. Gene Expr
Patterns. 2009;9:1–26.
104. Zhao L, Wang D, Wang Q, Rodal AA, and Zhang YQ.
Drosophila cyp regulates synaptic development and
endocytosis by suppressing lamentous actin assembly.
PLoS Genet. 2013;4.
105. Ng J, Luo L. Rho GTPases regulate axon growth through
convergent and divergent signaling pathways. Neuron.
2004;5:779–93.
106. Siudeja K, Grzeschik NA, Rana A, de Jong J, Sibon C.
Colin/Twinstar phosphorylation levels increase in re-
sponse to impaired coenzyme a metabolism. PLoSOne
2012;8:e43145.
107. Norris AD, Dyer JO, Lundquist EA. The Arp2/3 com-
plex, UNC-115/abLIM, and UNC-34/Enabled regulate
axon guidance and growth cone lopodia formation in
Caenorhabditis elegans. Neural Dev. 2009;4:38.
108. Struckhoff EC, Lundquist EA. The actin-binding protein
UNC-115 is an effector of Rac signaling during axon
pathnding in C. elegans. Development. 2003;130:693–
704.
109. Shakir MA, Gill JS, Lundquist EA. Interactions of UNC-
34 Enabled with RacGTPases and the NIK kinase MIG-
15 in Caenorhabditis elegans axon pathnding and neu-
ronal migration. Genetics. 2006;172:893–913.
110. Withee J, Galligan B, Hawkins N, Garriga G. Caenorhab-
ditis elegans WASP and Ena/VASP proteins play com-
pensatory roles in morphogenesis and neuronal cell mi-
gration. Genetics. 2004;167:1165–76.
111. Lebrand C, Dent EW, Strasser GA, Lanier LM, Krause
M, Svitkina TM, Borisy GG, Gertler FB. Critical role
of Ena/VASP proteins for lopodia formation in neu-
rons and in function downstream of netrin-1. Neuron.
2004;42:37–49.
112. Barzik M, Kotova TI, Higgs HN, Hazelwood L, Hanein
D, Gertler FB, Schafer DA. Ena/VASP proteins enhance
actin polymerization in the presence of barbed end cap-
ping proteins. J Biol Chem. 2005;280:28653–62.
113. Bear JE, Gertler FB. Ena/VASP: towards resolving
a pointed controversy at the barbed end. J Cell Sci.
2009;122:1947–53.
114. Applewhite DA, Barzik M, Kojima S, Svitkina TM,
Gertler FB, Borisy GG. Ena/VASP proteins have an anti-
capping independent function in lopodia formation.
Mol Biol Cell. 2007;18:2579–91.
115. Ans JE, Snow JJ, Gunnarson AL, Ou G, Stahlberg
H, McDonald KL, Scholey JM. Functional modula-
tion of IFT kinesins extends the sensory repertoire of
ciliated neurons in Caenorhabditis elegans. J Cell Biol.
2006;172:663–9.
116. Snow JJ, Ou G, Gunnarson AL, Walker MR, Zhou HM,
Brust MI, Scholey JM. Two anterograde intraagellar
transport motors cooperate to build sensory cilia on C.
elegans neurons. Nat Cell Biol. 2004;6:1109–1113.
117. Insinna C, Pathak N, Perkins B, Drummond I, Besharse
JC. The homodimeric kinesin, Kif17, is essential for
vertebrate photoreceptor sensory outer segment develop-
ment. Dev Biol. 2008;1:160–70.
118. Yoon BC, Zivraj KH, Strochlic L, Holt CE. 14-3-3 Pro-
teins regulate retinal axon growth by modulating ADF/
Colin activity. Dev Neurobiol. 2012;4:600–14.
119. Gurniak CB, Perlas E, Witke W. The actin depolymer-
izing factor n-colin is essential for neural tube mor-
phogenesis and neural crest cell migration. Dev Biol.
2005;1:231–41.
120. Huiping Z, James OEE, Chen M, Gary MS, Edward JL,
Richard HF. Association between CFL1 gene polymor-
phisms and spina bida risk in a California population.
BMC Medical Genetics. 2007;8:12.
121. Robertson SP, Twigg SR, Sutherland-Smith AJ, Bian-
calana V, Gorlin RJ, Horn D, Kenwrick SJ, Kim CA,
Morava E, Newbury-Ecob R, Orstavik KH, Quarrell
OW, Schwartz CE, Shears DJ, Suri M, Kendrick-Jones
J, Wilkie AO. Localized mutations in the gene encoding
the cytoskeletal protein laminA cause diverse malfor-
mations in humans. Nature Genetics. 2003;4:487–91.
122. Beetz C, Johnson A, Schuh AL, Thakur S, Varga RE,
Fothergill T, Hertel N, Bomba-Warczak E, Thiele H,
Nürnberg G, Altmüller J, Saxena R, Chapman ER, Dent
EW, Nürnberg P, Audhya A. Inhibition of TFG function
causes hereditary axon degeneration by impairing endo-
plasmic reticulum structure. 2013;13:5091–6.
123. Abdollahi MR, Morrison E, Sirey T, Molnar Z, Hayward
BE, Carr IM, Springell K, Woods CG, Ahmed M, Hat-
tingh L, Corry P, Pilz DT, Stoodley N, Crow Y, Taylor
GR, Bonthron DT, Sheridan E. Mutation of the variant
a-tubulin TUBA8 results in polymicrogyria with optic
nerve hypoplasia. Am J Human Genetics. 2009;85:737–
44.
124. Shu Y, Jennifer AF, Kelly LW, Sadaf TW, Roger P, Garth
AN, Ian PB. Mutation analysis and immunopathological
studies of PFN1 in familial and sporadic amyotrophic lat-
eral sclerosis. 2013;9: 2235:e7–10.
125. Thomas S, Encha-Razavi F, Devisme L, Etchevers H,
Critical ReviewsTM in Eukaryotic Gene Expression
Anand, Bammidi & Bali
268
Bessieres-Grattagliano B, Goudefroye G, Elkhartou N,
Pateau E, Ichkou A, Bonnière M, Marcorelle P, Parent
P, Manouvrier S, Holder M, Laquerrière A, Loeuillet L,
Roume J, Martinovic J, Mougou-Zerelli S, Gonzales M,
Meyer V, Wessner M, Feysot CB, Nitschke P, Leticee N,
Munnich A, Lyonnet S, Wookey P, Gyapay G, Foliguet
B, Vekemans M, Attié-Bitach T. High-throughput se-
quencing of a 4.1 Mb linkage interval reveals FLVCR2
deletions and mutations in lethal cerebral vasculopathy.
Human Mutation. 2010;10:1134–41.
... Genes meeting the criteria for hyperglycaemic memory were attributed to the cytoskeletal and nuclear compartments. The cytoskeleton has been shown to be involved in the pathogenesis of diabetic microvascular complications [8,9]. More importantly, sustained changes in nuclear factors have been reported to have a large-scale impact on gene expression, resulting in changes in antioxidative defence mechanisms to hyperglycaemic stress [10]. ...
Article
Aims/hypothesis: The aim of this study was to evaluate damage to the neurovascular unit in a mouse model of hyperglycaemic memory. Methods: A streptozotocin-induced mouse model of diabetes (C57BL/6J background) received insulin-releasing pellets and pancreatic islet-cell transplantation. Damage to the neurovascular unit was studied by quantitative retinal morphometry for microvascular changes and microarray analysis, with subsequent functional annotation clustering, for changes of the retinal genome. Results: Sustained microvascular damage was confirmed by persistent loss of pericytes in the retinal vasculature (PC/mm2): compared with healthy controls (1981 ± 404 PC/mm2), the pericyte coverage of the retinal vasculature was significantly reduced in diabetic mice (1571 ± 383 PC/mm2, p < 0.001) and transplanted mice (1606 ± 268 PC/mm2, p < 0.001). Genes meeting the criteria for hyperglycaemic memory were attributed to the cytoskeletal and nuclear cell compartments of the neurovascular unit. The most prominent regulated genes in the cytoskeletal compartment were Ddx51, Fgd4, Pdlim7, Utp23, Cep57, Csrp3, Eml5, Fhl3, Map1a, Mapk1ip1, Mnda, Neil2, Parp2, Myl12b, Dynll1, Stag3 and Sntg2, and in the nuclear compartment were Ddx51, Utp23, Mnda, Kmt2e, Nr6a1, Parp2, Cdk8, Srsf1 and Zfp326. Conclusions/interpretation: We demonstrated that changes in gene expression and microvascular damage persist after euglycaemic re-entry, indicating memory. Data availability: The datasets generated during and/or analysed during the current study are available in the GEO repository, GSE87433, www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=idmbysgctluxviv&acc=GSE87433 .
Article
Full-text available
Physical activity of respiratory and olfactory cilia of animals and humans in the context of L.A. Blumenfeld concept, according to which protein molecules are “machines that perform chemical trans-formation” and create “a meaningful order”, has been investigated and analyzed.
Article
Full-text available
Monocular deprivation (MD) during the critical period (CP) shifts ocular dominance (OD) of cortical responsiveness toward the nondeprived eye. The synaptic mechanisms underlying MD-induced OD plasticity, in particular the contribution of cortical inhibition to the plasticity, have remained unsolved. In this study, using in vivo whole-cell voltage-clamp recordings, we revealed eye-specific excitatory and inhibitory synaptic inputs to layer 4 excitatory neurons in mouse primary visual cortex (V1) at a developmental stage close to the end of CP. We found in normally reared mice that ocular preference is primarily determined by the contralateral bias of excitatory input and that inhibition does not play an active role in shaping OD. MD results in a parallel reduction of excitation and inhibition driven by the deprived eye, while reducing the inhibition but preserving the excitation driven by the nondeprived eye. MD of longer periods causes larger changes in synaptic amplitude than MD of shorter periods. Furthermore, MD resulted in a shortening of onset latencies of synaptic inputs activated by both contralateral and ipsilateral eye stimulation, while the relative temporal relationship between excitation and inhibition driven by the same eye was not significantly affected. Our results suggest that OD plasticity is largely attributed to a reduction of feedforward input representing the deprived eye, and that an unexpected weakening of cortical inhibitory connections accounts for the increased responsiveness to the nondeprived eye.
Article
Full-text available
Author Summary Synapses are specialized junctions at which neurons communicate with target cells. To establish properly wired neuronal circuits, synapses must grow in size and strength with a high degree of accuracy. The actin cytoskeleton plays a crucial role in the formation and function of synapses, but the underlying mechanisms remain poorly understood. The Drosophila neuromuscular junction (NMJ) is an excellent model for studying synaptic development and function. By analyzing Drosophila mutants of the cytoplasmic FMRP interacting protein Cyfip, we establish that this protein inhibits the assembly of filamentous actin (F-actin). At cyfip mutant NMJ synapses, F-actin assembly was accelerated and NMJ terminals were shorter and grew supernumerary buds. Furthermore, neurotransmission was not sustained under high-frequency stimulation. These changes could be caused by defects in synaptic endocytosis, which would compromise the endocytic attenuation of signaling pathways such as the NMJ growth-promoting bone morphogenetic protein (BMP) pathway. Indeed, BMP signaling was upregulated in cyfip mutants. We propose that Cyfip regulates synaptic development and function by inhibiting F-actin assembly, which in turn downregulates BMP signaling via endocytosis. This study establishes a novel role for Cyfip-mediated regulation of the actin cytoskeleton at the Drosophila NMJ.
Article
The vertebrate retina contains seven major neuronal and glial cell types in an interconnected network that collects, processes and sends visual signals through the optic nerve to the brain, Retinal neuron differentiation is thought to require both intrinsic and extrinsic factors, yet few intrinsic gene products have been identified that direct this process. Math5 (Atoh7) encodes a basic helix-loop-helix (bHLH) transcription factor that is specifically expressed by mouse retinal progenitors. Math5 is highly homologous to atonal, which is critically required for R8 neuron formation during Drosophila eye development. Like R8 cells in the fly eye, retinal ganglion cells (RGCs) are the first neurons in the vertebrate eye. Here we show that Math5 mutant mice are fully viable, yet lack RGCs and optic nerves. Thus, two evolutionarily diverse eye types require atonal gene family function for the earliest stages of retinal neuron formation. At the same time, the abundance of cone photoreceptors is significantly increased in Math5(-/-) retinae, suggesting a binary change in cell fate from RGCs to cones. A small number of nascent RGCs are detected during embryogenesis, but these fail to develop further, suggesting that committed RGCs may also require Math5 function.
Article
In previous work we characterized a brain derived collapsing factor that induces the collapse of dorsal root ganglion growth cones in culture (Raper and Kapfhammer, 1990). To determine how the growth cone cytoskeleton is rearranged during collapse, we have compared the distributions of F-actin and microtubules in normal and partially collapsed growth cones. The relative concentration of F-actin as compared to all proteins can be measured in growth cones by rationing the intensity of rhodamine-phalloidin staining of F-actin to the intensity of a general protein stain. The relative concentration of F-actin is decreased by about one half in growth cones exposed to collapsing factor for five minutes, a time at which they are just beginning to collapse. During this period the relative concentration of F-actin in the leading edges of growth cones decreases dramatically while the concentration of F-actin in the centers decreases little. These results suggest that collapse is associated with a net loss of F-actin at the leading edge. The distributions of microtubules in normal and collapsing factor treated growth cones were examined with antibodies to tyrosinated and detyrosinated isoforms of alpha-tubulin. The tyrosinated form is found in newly polymerized microtubules while the detyrosinated form is not. The relative proximal-distal distributions of these isoforms are not altered during collapse, suggesting that rates of microtubule polymerization and depolymerization are not greatly affected by the presence of collapsing factor. An analysis of the distributions of microtubules before and after collapse suggests that microtubules are rearranged, but their polymerization state is unaffected during collapse. These results are consistent with the hypothesis that the brain derived collapsing factor has little effect on microtubule polymerization or depolymerization. Instead it appears to induce a net loss of F-actin at the leading edge of the growth cone.
Article
Mutations in PFN1, a gene encoding the actin monomer-binding protein profilin 1, were recently reported in 1% to 2% of familial amyotrophic lateral sclerosis (ALS) patients. In vitro functional studies suggested that PFN1 mutations lead to ubiquitin-positive inclusions and impairment of cytoskeletal pathways. In the present study, mutation analysis of PFN1 was performed in an Australian cohort of 110 ALS families and 715 sporadic ALS patients. No PFN1 mutations were identified in familial ALS patients. Two rare non-synonymous variants (E117D and E117G) were found in sporadic ALS patients at similar incidences to that reported in public SNP databases. Immunostaining of PFN1 in sporadic ALS and familial ALS patients, including those with mutations in SOD1, FUS, UBQLN2 and C9ORF72, found no PFN1-positive inclusions in spinal motor neurons. Our data suggest that PFN1 mutations and pathology are not common in an Australian ALS cohort of predominantly European ancestry.
Article
Formins are actin binding proteins conserved across species from plants to humans. The formin family is defined by their common FH2 domains. The 15 distinct human formins are involved in a broad range of cellular functions, including cell adhesion, cytokinesis, cell polarity, and cell morphogenesis. Their commonality is actin polymerization activity inherent to FH2 domains. While still requiring much study, biochemical activity of formins has been carefully described. In contrast, much less is known of their activities in complex living systems. With the diversity of the formin family and the actin structures that they affect, an extensive future of study beckons. In this study, we report the expression level of all 15 formins in 22 different human cell and tissue types using quantitative real-time PCR (qPCR). Identification of major themes in formin expression and documentation of expression profiles should facilitate the cellular study of formins. © 2013 Wiley Periodicals, Inc.
Article
Hereditary spastic paraplegias are a clinically and genetically heterogeneous group of gait disorders. Their pathological hallmark is a length-dependent distal axonopathy of nerve fibers in the corticospinal tract. Involvement of other neurons can cause additional neurological symptoms, which define a diverse set of complex hereditary spastic paraplegias. We present two siblings who have the unusual combination of early-onset spastic paraplegia, optic atrophy, and neuropathy. Genome-wide SNP-typing, linkage analysis, and exome sequencing revealed a homozygous c.316C>T (p.R106C) variant in the Trk-fused gene (TFG) as the only plausible mutation. Biochemical characterization of the mutant protein demonstrated a defect in its ability to self-assemble into an oligomeric complex, which is critical for normal TFG function. In cell lines, TFG inhibition slows protein secretion from the endoplasmic reticulum (ER) and alters ER morphology, disrupting organization of peripheral ER tubules and causing collapse of the ER network onto the underlying microtubule cytoskeleton. The present study provides a unique link between altered ER architecture and neurodegeneration.
Article
The pigmented ciliary epithelium (PCE) of mammalian eye harbors resident population of stem cells that lie in apposition with endothelial cells which release vascular endothelial growth factor (VEGF) that may influence the fate and function of these stem cells in ways that remain unclear. We examined the role of VEGF in proliferation of PCE stem cells and expression of Notch, Jagged, N-Cadherin and β- Catenin which are known to maintain proliferation state of neural stem cells. We cultured human PCE cells obtained from 12-20 weeks old fetal eyes. The neurospheres were analyzed for the proliferation capacity of PCE stem cells in presence of VEGF on 3,6 and 9 day. Real time PCR was used to quantitate the mRNA expression of above mentioned genes on PCE derived neurospheres on 3,6 and 9 day. We found increased number of neurospheres when PCE stem cells were stimulated with VEGF alongwith epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) than EGF and bFGF. BrdU immunostaining was done to analyze the proliferation of CE cells and presence of neural and retinal progenitor markers such as Nestin and Pax6 were also investigated. An increased Notch and Jagged mRNA was observed on 6th day in VEGF, EGF and bFGF treated PCE cells as compared to 0,3 and 9 day. A similar pattern was noticed with N-cadherin and β-catenin mRNA levels. These findings may clarify the role of VEGF on PCE stem cell proliferation with possible involvement of Notch, Jagged, N19 cadherin and β-Catenin. The data may suggest importance of harvesting 6th day neurospheres for transplantation purposes in preclinical investigations pertaining to retinal degenerative diseases, however, additional studies are needed to substantiate the findings.