You are on page 1of 7

Biochimie 107 (2014) 51e57

Contents lists available at ScienceDirect

Biochimie
journal homepage: www.elsevier.com/locate/biochi

Mini-review

Phosphatidic acid in neuronal development: A node for membrane


and cytoskeleton rearrangements
Mohamed-Raafet Ammar, Nawal Kassas, Marie-France Bader, Nicolas Vitale*
Institut des Neurosciences Cellulaires et Int
egratives (INCI), UPR-3212 Centre National de la Recherche Scientique & Universit
e de Strasbourg,
5 rue Blaise Pascal, 67084 Strasbourg, France

a r t i c l e i n f o

a b s t r a c t

Article history:
Received 21 May 2014
Accepted 30 July 2014
Available online 8 August 2014

Phosphatidic acid (PA) is the simplest phospholipid naturally existing in all-living organisms. It constitutes only a minor fraction of the total cell lipids but has attracted considerable attention being both a
lipid second messenger and a modulator of membrane shape. The pleiotropic functions of PA are the
direct consequence of its very simple chemical structure consisting of only two acyl chains linked by
ester bonds to two adjacent hydroxyl groups of glycerol, whose remaining hydroxyl group is esteried
with a phosphomonoester group. Hence the small phosphate head group of PA gives it the shape of a
cone providing exibility and negative curvatures in the context of a lipid bilayer. In addition, the
negatively charged phosphomonoester headgroup of PA is unique because it can potentially carry one or
two negative charges playing a role in the recruitment of positively charged molecules to biomembranes.
In consequence, PA has been proposed to play various key cellular functions. In the brain, a ne balance
between cell growth, migration and differentiation, and cell death is required to sculpt the nervous
system during development. In this review, we will summarize the various functions that have been
proposed for PA in neuronal development.
te
 franaise de biochimie et biologie Mole
culaire (SFBBM). All rights
2014 Elsevier B.V. and Socie
reserved.

Keywords:
Phosphatidic acid
Neurite outgrowth
Phospholipase D
Dendritic spine
Vesicular fusion

1. Introduction
During brain development, neurons exhibit various morphological changes including axon and dendrite outgrowth, dendritic
branching and spine formation, as well as synapse formation,
which are critical events in the establishment of neuronal networks. Because of its complexity and heterogeneity, most molecular aspects of the nervous system development have been studied
using cell culture models. These include various types of neurons,
but also the rat neural crest derivative PC12 cells that develop
neuronal features in response to neuronal growth factor (NGF)
treatment.
In culture, hippocampal neurons undergo morphological
changes that were rst described by Dotti et al. [1]. Shortly after
plating, the cells begin to develop lamellipodia around the cell body
(Stage 1). The second phase of development is marked by the loss of
lamellipodia and the extension of neurites or neurite outgrowth
(Stage 2). After 1e2 days in vitro, the majority of cells have an

* Corresponding author.
E-mail address: vitalen@unistra.fr (N. Vitale).

asymmetrical morphology characterized by a single and long


neurite, which will become the axon, and several extensions
shorter than the axon, which will become dendrites (Stage 3 and 4).
The last stage (Stage 5) represents the maturation of the axonal and
dendritic branching, including dendritic spine formation and synaptogenesis [2,3].
Morphological changes of neurons during development are
essentially based on two different cellular mechanisms: i) the
reorganization of the cytoskeleton [4] and ii) the enlargement of
the cell surface sustained by exocytosis of cytoplasmic vesicles [5].
At the tips of the growing neurites, growth cones are specialized
highly mobile cellular compartments consisting of a central region
lled with organelles and microtubules and a dynamic peripheral
region rich in actin. Growth cone morphology, guidance and
motility are driven by cyclical polymerization and depolymerization of actin laments, whereas the structure of the
axon and its elongation rely on microtubules [4,6e8]. Cytoskeletal
dynamics also play a key role in dendritic spine formation and
maturation. Dendritic spines are small protrusions emerging from
parent dendrites that contain postsynaptic structures such as
postsynaptic density, actin laments and microtubules (Reviewed
in Ref. [9]). In vivo, spines represent the postsynaptic zones that

http://dx.doi.org/10.1016/j.biochi.2014.07.026
 te
 franaise de biochimie et biologie Mole
culaire (SFBBM). All rights reserved.
0300-9084/ 2014 Elsevier B.V. and Socie

52

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57

establish the physical contact with the presynaptic area to form


functional synapses that allow the chemical transfer of signal between two neurons.
Morphological maturation of neurons is also accompanied by
changes in the membrane surface provided by two membrane
trafcking mechanisms: exocytosis and endocytosis. These two
mechanisms are highly regulated and provide directional transportation of intracellular membrane components [10]. Indeed,
axons or neurites extend sometimes over long distances to reach
their synaptic targets. Their elongation requires the increase in
membrane surface, which is supported by anterograde vesicular
transport from the cell body to the growth cone followed by
membrane fusion/exocytosis to ensure the addition of vesicular
membrane and surface expansion at the growth cone [5,11,12]. In
contrast, endocytosis and the reduction of cell surface is involved in
neurite retraction and growth cone collapse [13e15].
Phosphatidic acid (PA) is a pleiotropic lipid that plays an
important structural role as a precursor of various glycerophospholipids. PA is also considered as a key player in the transmission, amplication and regulation of a variety of intracellular
signaling and cellular functions, such as cell proliferation, vesicle/
membrane trafcking and cytoskeletal organization. At the molecular level, PA interacts with various proteins to modulate catalytic activity and/or membrane association [16,17], and these
include GTPases, kinases, phosphatases, nucleotide binding proteins and phospholipases [18]. Many PA partners are involved in the
regulation of actin dynamics and membrane trafcking and some of
them have been involved in neuronal development. In this review,
we will summarize the current knowledge linking PA to the
cellular/molecular mechanisms that govern neuronal development.
2. Synthesis and degradation pathways for PA
Within cells, PA occurs as a structural intermediate or as a
signaling molecule. In mammals, structural PA is synthesized
through two acylation reactions: the glycerol 3-phosphate (G-3-P)
pathway and the dihydroxyacetone phosphate (DHAP) pathway
(Fig. 1). These reactions catalyzed by the glycerol-3-P acyltransferase (G-3-P AT) and the dihydroxyacetone phosphate acyltransferase
(DHAP AT) produce a monoacylated form of PA, the lysoPA (LPA)
transformed in a second acylation step into PA [19]. PA generated
from these de novo pathways is a key intermediate in the synthesis
of all glycerophospholipids and triacyglycerols. This structural PA is
rapidly converted by phosphatidic acid phosphohydrolase (PAP) to

Fig. 1. Biosynthetic pathways of structural and signaling PA. PA is a central phospholipid for biosynthetic and signaling reactions. Red arrows denote biosynthetic reactions that lead to structural PA synthesis whereas formation of signaling pools of PA
is shown with blue arrows.

diacyglycerol (DAG), which is an important source for the biosynthesis of triacylglycerols and phospholipids including phosphatidylcholine
(PC),
phosohatidylethanolamine
(PE)
and
phosphatidylserine (PS) [20]. In addition, PA can be deacylated by
phospholipases A (PLA) to LPA or degraded by CDP-DAG synthetase
into CDP-diacylglycerol, which is responsible for the synthesis of
phosphatidylinositol (PI), phosphatidylglycerol (PG) and cardiolipin (CL) [19] (Fig. 1).
Signaling forms of PA can be formed by three alternative
biosynthetic pathways (Fig. 1). The rst pathway involves phospholipase D (PLD), which catalyzes the hydrolysis of the distal
phosphodiester bond in PC to form PA and choline. PLD preferentially catalyzes a transphosphatidylation reaction to produce
phosphatidylalcohols at the expense of PA production in the presence of primary alcohols, such as ethanol and 1-butanol. Measuring
the production of labeled phosphatidylalcohols has been until
recently an important method used to study cellular functions of
PLD [21,22]. Cloning and characterization of genes encoding PLD
proteins have identied at rst two isoforms (PLD1 and PLD2) in
animal cells. Two alternative splicing variants of PLD1 (PLD1a and
PLD1b) and three for PLD2 (PLD2a, PLD2b and PLD2c) were subsequently identied. Both PLD isoforms contain two HKD motifs,
which are critical for enzymatic catalysis, a phox (PX) domain, a
pleckstrin homology (PH) domain and a PIP2 binding domain [23].
In addition, PLD1 contains a loop domain that is not found in
PLD2 and seems to be involved in the auto-inhibition of PLD1
[24,25]. PLD1 which is characterized by low basal activity, is activated principally by three classes of proteins: ARF GTPases, Rho
GTPases and several members of the family of protein kinases
including PKCs and RSK2. PLD2 which exhibits a higher basal activity, is also modulated by ARF and Rac proteins [26e28]. More
recently, structurally different isoforms of PLD were identied.
These include PLD3, PLD4 and a mitochondrial PLD (mito-PLD).
Mito-PLD in contrast to classical mammalian PLDs uses cardiolpin
as substrate to generate PA. An important structural difference
between mito-PLD and PLD1/2 is that mito-PLD has only a single
HKD half-catalytic site, requiring it to dimerize to create an active
enzymatic complex. Furthermore, mito-PLD displays no PX, PH or
PIP2 membrane binding domains and needs its N-terminal
sequence for mitochondrial surface anchoring. Regarding PLD3 and
PLD4 proteins, although their catalytic activity has not been
established to date PLD3 appears to be involved in cell differentiation [29] and PLD4 in phagocytosis of microglial cells [30].
The phosphorylation of DAG using ATP as a phosphate source by
DAG kinase (DGK) produces PA. Ten isoforms for mammalians DGK
(a, b, g, d, h, k, , z, i, and q) have been cloned and characterized.
These isoforms are classied in ve subtypes and all of them have a
conserved catalytic domain and at least two cysteine rich domains.
The three type I DGKs (a, b and g) contain calcium-binding EF
hands motifs in their N-termini, while type II (d, h and k) have
pleckstrin homology (PH) domains at their N termini. The type III
DGK () has the simplest structure and does not contain regulatory
units. The types IV DGK (z and i) have ankyrin repeats at their C
termini and a region homologous to the MARCKS protein's phosphorylation site. Finally, DGK q denes type V, has three cysteinerich domains and a PH domain [31].
The acylation of LPA by lysoPA-acyltransferases (LPAAT) also
leads to PA formation [32]. Six human LPAAT isoforms have been
cloned and characterized [33]. The isoforms LPAAT 1 & 2 have 34%
sequence homology and the highest LPAAT activity [34]. These
isoforms also contain from two to four transmembrane domains
and two highly conserved motifs (H (X) 4D and EGT), which are
essential for catalytic activity of the family of acyltransferases
[35,36]. Interestingly, more proteins displaying LPAAT activities
have been recently identied. For instance the protein RIBEYE in

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57

synaptic ribbon was shown to possess LPAAT activity and thus to be


a physiologically relevant source of PA which may facilitate synaptic
vesicle trafcking [37].
The complexity and diversity of PLD, DGK and LPAAT families
suggest that they are involved in various specic cellular functions
and are probably not redundant. In general, signaling PA generated
from these enzymes is implicated in multiple cell functions such as
cytoskeleton organization, cell survival and proliferation, membrane and vesicle trafcking.
3. PA in neurite outgrowth
The rst report suggesting a link between PA synthesis and
neuronal development came from a study focusing on phospholipid metabolism in NGF-treated PC12 cells [38]. Since then, PA
generated from different enzymatic pathways has been suggested
to be involved in neurite outgrowth. For instance, several groups
have reported that PLD1 and PLD2 isoforms contribute to neurite
outgrowth through the production of PA (reviewed in Ref. [39]).
DGKb/z isoforms were also shown to promote neurite and dendritic
extension in a PA-dependent manner [40,41].
Despite these original observations, however, little is currently
known on how PA production promotes neural development. PA via
its numerous downstream effectors [16,17] could act as a switch-on
molecule and/or as a coordinator of signals controlling actin dynamics and membrane trafcking.
3.1. PA and actin dynamics
PA is linked to actin dynamics via two large protein families: the
Rho family of GTPases and several protein kinase families known to
modulate actin cytoskeleton organization.
The Rho family of GTPases and related molecules play an
important role in various aspects of neuronal development,
including neurite outgrowth and differentiation, axonal pathnding and dendritic spine formation and maintenance
(Reviewed in Ref. [42]). Most of these effects are orchestrated
through the cytoskeleton since the Rho family GTPases, RhoA, Rac1
and Cdc42 has been shown to regulate a variety of cytoskeletal
dynamics [43]. Recent studies indicate that signaling PA generated
by both PLD and DGK plays critical roles in regulating the activity of
some members of Rho family of GTPases either by modulating their
membrane localization or by acting on their regulatory proteins
[44,45]. For instance, the guanine nucleotide-exchange factors
(GEFs) including Dock1 and Dock2 [46,47] and the GTPase activating proteins (GAPs) RA-RhoGAP [48] are directly sensitive to PA.
Interestingly, it was recently shown that DGKb-generated PA binds
to the PH domain of RA-RhoGAP, enhances its GAP activity toward
RhoA and promotes RA-RhoGAP-mediated neurite extension [48].
As well, PLD-mediated PA, through an interaction with the polybasic amino acid cluster of Dock2, seems to be required to recruit
cytosolic Dock to the plasma membrane, to subsequently activate
Rac and mediate local reorganization of the actin cytoskeleton [47].
Three kinase families are known for being activated by PA. PAK1
kinase is a target for PA produced by DGKz [49]. In neurons, PAK
isoforms have been shown to regulate various aspects of neuronal
development, including neuronal polarization and migration,
neurite outgrowth and dendritic spine formation [50e52]. PAK1
can trigger actin cytoskeleton reorganization through various
pathways implicating either the actin motor protein myosin II [53],
the actin depolymerizing LIM Kinase (LIMK) [54], the actin nucleation protein Arp2/3 [55] or the dissociation of Rac1 from RhoGDI
and its activation [49].
PA activates also the phospholipid kinase phosphatidylinositol
4-phosphate 5-kinase (PIP5K) [32,56e58], which catalyzes the

53

phosphorylation of the membrane phospholipid phosphatidylinositol 4-phosphate to generate the pleiotropic PIP2. The PA pool
involved in the activation of PIP5K has not been precisely characterized but it seems that the two major sources of signaling PA,
namely PLD and DGK, are involved [58e61]. The three mammalian
PIP5K isozymes, a, b, and g, have been described as playing a role in
actin cytoskeleton reorganization (Reviewed in Ref. [62]) and
linked to neuronal outgrowth, axon elongation and branching, and
growth cone morphology [63e65].
Finally, PA has been described to regulate the membrane localization and activity of PKC isoforms a, and z [66e68]. PKCs are
known to inuence the morphology of the actin lament cytoskeleton. This kinase family has been implicated in a variety of
cellular functions, including neurite outgrowth (reviewed in Ref.
[69]).
In addition to GTPase and kinase regulated pathways, PA has
been reported to inuence the actin organization by interacting
directly with some actin-binding proteins. For instance, PA interacts with vinculin [70,71], an actin-binding protein described to
play a role in neurite outgrowth in PC12 cells and neurons. Yet, actin
plays intricate functions in neurite elongation, which range from a
structural function and the organization of vesicular trafcking to
the establishment/stabilization of cell membrane domains dedicated to vesicular fusion and membrane incorporation (Fig. 2).
3.2. PA and membrane trafcking
PA has been proposed to play a role in various trafcking events,
including endocytosis and regulated exocytosis leading to the
release of hormones from endocrine and neuroendocrine cells,
neurotransmitters from neurons, IL-8 from epithelial cells, mast cell
degranulation and insulin-stimulated translocation of the glucose
transporter Glut4 to the cell membrane (reviewed in Refs. [72,73].
These observations have been obtained mostly by the use of molecular and pharmacological inhibitors of PLDs and suggest that
PLD1 is the isoform mostly involved in membrane trafcking,
although few examples for the involvement of DGK-generated PA in
membrane trafcking have been also described.
Evidence supporting the involvement of PLD1-generated PA in
the late steps of exocytosis, such as membrane fusion and/or pore
expansion comes from studies in chromafn cells in which injection of a dominant-negative PLD1 mutant [74] or PLD1 silencing
[75] affects the fusion rate of single secretory granule release. Yet,
the mechanism(s) by which PA promotes membrane fusion remains debated. It may be linked to its ability to generate membrane
curvature and facilitate fusion [75], modulate syntaxin-1 activity
and the docking of vesicles [76] and/or recruit key proteins required
for the fusion process.
Although PLD-generated PA has been shown to be crucial to
neurite outgrowth in a variety of neuronal cell models (reviewed in
Refs. [39], the mechanisms implicating PA in the underlying
membrane trafcking events remain elusive. Supporting the
importance of PLD1-generated PA in neurite outgrowth, we
recently observed that neuronal maturation was signicantly
delayed in Pld1 knockout neurons [77]. Interestingly, this alteration
of neuronal development very much recapitulated that observed in
Rsk2 knockout neurons, a model for the human X-linked CofnLowry syndrome, a severe mental retardation disease [77]. We
found also that PLD1 became phosphorylated and activated by the
kinase RSK2 during neurite outgrowth in PC12 cells [77]. PLD1
activation leads to an increase of PA levels at the tips of growing
neurites as revealed by the PA sensor Spo20p-GFP [77]. Interestingly, PLD1 was found to be associated to trans Golgi-derived vesicular structures, also positive for the vesicular SNARE VAMP-7/
TiVAMP. These vesicles moved anterogradly and retrogradly in

54

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57

Fig. 2. PA plays a role in axon and dendrite outgrowth, dendritic branching and spine formation. Schematic representation of dendritic spines and a growth cone with microtubules
(green), actin cytoskeleton (red) and trafcking vesicles. In dendrites, elevated PA favors spine maintenance through the regulation of actin dynamics. In growth cones, PA is
involved in neurite outgrowth by controlling actin organization and vesicular trafcking for membrane supply. The uorescence images on the right-hand side show a cortical
neuron dendrite labeled to visualize actin and MAP2 and a growth cone with its tubulin and actin cytoskeleton. Bars 10 mm.

the developing neurites [77]. RSK2 and PLD1 were found to interact
at the tip end of growing neurites, in line with the increased PA
level at this location [77]. We monitored the fusion rate of PLD1/
VAMP-7 vesicles and found that RSK2 and PLD1 inhibitors
dramatically reduced membrane supply required for neurite
outgrowth [77]. Taken together, these results reveal for the rst
time the direct involvement of PA-mediated membrane trafcking
in neurite outgrowth. The observation that quenching PA by
expressing a PA sensor in PC12 cells strongly reduces the number
and length of the NGF-induced neurites [77] suggests that PA acts
in part by recruiting key proteins at the vesicle fusion site during
neurite outgrowth.
4. PA in dendritic arborization and spine regulation
Pld1 knockout affects dendritic branching. Specically, the
number of secondary branching dendrites is signicantly reduced
in neurons cultured from Pld1-/- mice [77]. Additionally, the density of the spines, the tiny protrusions (0.5 mm wide, 2 mm long) on
neuronal dendrites that receive the majority of excitatory synaptic
inputs, is signicantly reduced in Pld1-/- hippocampal neurons and
this reduction in spine density specically affects mushroom and
branched spines, which are the forms of mature spines [77].
Accordingly, a previous report showed that the GTPase RalB promotes branching through a pathway involving PLD [78]. PLD1 has
also been described to negatively regulate dendritic branching in
hippocampal neurons [79], but this apparent contradiction is most
likely related to the methods used to modify PLD1 expression
levels, i.e. overexpression or silencing versus genetically knocked

down PLD1. Anyhow, these results imply critical functions of PLD1produced signaling PA in early stages of neuronal growth and
development.
Additionally, dendrite arborization and spines seem to rely on
DGK-generated signaling PA. For instance, overexpression of wildtype DGKb promotes dendrite outgrowth and spine maturation in
transfected hippocampal neurons, whereas a kinase-dead mutant
DGK has no effect [40]. Accordingly, primary cultured hippocampal
neurons from DGKb KO mice had less branches and spines
compared to the wild type [41]. DGKz has also been reported to be
important for the maintenance of dendritic spines and the regulation of the PA/DAG balance at excitatory synapses [80,81]. Indeed
the C-terminal PDZ-binding motif of DGKz interacts with the PDZ
domains of the postsynaptic density protein PSD-95, an abundant
postsynaptic scaffolding protein that regulates excitatory synaptic
structure and function. This interaction promotes synaptic localization of DGKz at synapses. Functionally, DGKz overexpression increases spine density in cultured hippocampal neurons in a manner
that requires DGK catalytic activity but also PSD-95 binding,
whereas knockdown of DGKz decreases spine density. Mice decient for DGKz expression show a reduction in spine density in
hippocampal CA1 pyramidal neurons and exhibit reduced excitatory synaptic transmission. Time-lapse imaging in DGKz-decient
mice revealed impaired spine maintenance but not formation [80].
Taken together, these data also reveal a key role of DGK-generated
PA in dendritic ramication and spine formation and maturation.
By tightly controlling the conversion of DAG to PA, DGK either
stabilizes (PA-dependent) or destabilizes (DAG-dependent) dendritic spines (Fig. 2; recently reviewed in Ref. [87]).

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57

How may PA production promote and/or maintain dendritic


spines? Several PA effectors linked to actin dynamics play a role in
neurite outgrowth and have been also implicated in dendritic spine
formation (Fig. 2). For instance, a positive role for the serine/threonine kinase PAK1 in the regulation of dendritic spines has been
established by several studies [52,82e84]. Actin-regulatory proteins including prolin, gelsolin, a-actinin, vinculin, and the WiskotteAldrich syndrome protein-Arp2/3 complex have been shown
to regulate dendritic spines [85e89]. Most of them are activated by
PIP2 generated by PIP5Ka, a kinase activated by PA [90] and
forming a complex with DGKz [60,91]. PKCz, another notable PA
effector regulates dendritic spines [92].
Other PA effectors, like protein phosphatase 1 (PP1) [93] highly
enriched in postsynaptic densities in the brain [94], or the
mammalian target of rapamycin mTOR implicated in the regulation
of synaptic plasticity and memory [95], seem to be specically
involved in spine morphogenesis and maturation [96e98].
5. Conclusion
Because PA has both signaling functions and potential effects on
membrane topology, it is likely that its synthesis is tightly regulated
at various subcellular sites. One way the cell might achieve this is by
targeting the synthesizing enzymes DGK, PLD and LPAAT at precise
locations in neurons. For instance, we have shown that PLD1 is
found associated with the VAMP-7 vesicles opening the possibility
that PA synthesis occurs precisely at the site of vesicular fusion for
growth cone extension [77]. It is also conceivable that PA producing- and PA effector proteins might form multi-protein complexes
that spatially and temporally organize PA signaling pathways. The
development of PA sensors designed by fusing identied protein PA
binding domains with uorescent tags [99], will be an asset to
examine PA dynamics at the various stages of neuronal life.
Yet another aspect of PA that has not been investigated with care
is the inuence of the fatty-acyl chain composition on the functions
of PA. For instance, do the various PAs displaying saturated, mono-,
bi- or polyunsaturated fatty acids have similar effects on membrane
topology or recruit the same proteins? These questions are especially relevant in the context of neuronal development and
degeneration, since many studies have highlighted key functions of
dietary lipids in human cognitive activity [100]. The numerous
genetic models now available, the recent progress in lipidomics and
some new inhibitors will be helpful tools to clarify these intriguing
questions. Indeed, mice knockout for PLD1 or PLD2 showed early
brain development defects [101], whereas levels of specic species
of PA were altered in PLD2 knockout brain [102], illustrating the
notion that DGK and PLD isoforms may control the synthesis of
specic molecular pools of PA that could each contribute to various
aspects of neuronal development.
Conict of interest
The authors declare no conict of interest.
References
[1] C.G. Dotti, C.A. Sullivan, G.A. Banker, The establishment of polarity by hippocampal neurons in culture, J. Neurosci. Off. J. Soc. Neurosci. 8 (1988)
1454e1468.
[2] T.L. Fletcher, P. Cameron, P. De Camilli, G. Banker, The distribution of synapsin I and synaptophysin in hippocampal neurons developing in culture,
J. Neurosci. Off. J. Soc. Neurosci. 11 (1991) 1617e1626.
[3] A.M. Craig, R.J. Wyborski, G. Banker, Preferential addition of newly synthesized membrane protein at axonal growth cones, Nature 375 (1995)
592e594.
[4] E.W. Dent, S.L. Gupton, F.B. Gertler, The growth cone cytoskeleton in axon
outgrowth and guidance, Cold Spring Harb. Perspect. Biol. 3 (2011).

55

[5] K.H. Pfenninger, L. Laurino, D. Peretti, X. Wang, S. Rosso, G. Morni,


A. Caceres, S. Quiroga, Regulation of membrane expansion at the nerve
growth cone, J. Cell. Sci. 116 (2003) 1209e1217.
[6] L.A. Lowery, D. Van Vactor, The trip of the tip: understanding the growth
cone machinery, Nat. Rev. Mol. Cell. Biol. 10 (2009) 332e343.
[7] E.W. Dent, F.B. Gertler, Cytoskeletal dynamics and transport in growth cone
motility and axon guidance, Neuron 40 (2003) 209e227.
[8] K. Kalil, E.W. Dent, Touch and go: guidance cues signal to the growth cone
cytoskeleton, Curr. Opin. Neurobiol. 15 (2005) 521e526.
[9] T. Shirao, C. Gonzalez-Billault, Actin laments and microtubules in dendritic
spines, J. Neurochem. 126 (2013) 155e164.
[10] S.B. Sann, Growth cone stop signals: inviting to stay or sending away?
J. Neurosci. Off. J. Soc. Neurosci. 26 (2006) 8879e8880.
[11] Y. Chernyshova, I. Leshchyns'ka, S.C. Hsu, M. Schachner, V. Sytnyk, The neural
cell adhesion molecule promotes FGFR-dependent phosphorylation and
membrane targeting of the exocyst complex to induce exocytosis in growth
cones, J. Neurosci. Off. J. Soc. Neurosci. 31 (2011) 3522e3535.
[12] S.L. Sabo, A.K. McAllister, Mobility and cycling of synaptic protein-containing
vesicles in axonal growth cone lopodia, Nat. Neurosci. 6 (2003) 1264e1269.
[13] A.E. Fournier, F. Nakamura, S. Kawamoto, Y. Goshima, R.G. Kalb,
S.M. Strittmatter, Semaphorin3A enhances endocytosis at sites of receptor-Factin colocalization during growth cone collapse, J. Cell. Biol. 149 (2000)
411e422.
[14] W.M. Jurney, G. Gallo, P.C. Letourneau, S.C. McLoon, Rac1-mediated endocytosis during ephrin-A2- and semaphorin 3A-induced growth cone
collapse, J. Neurosci. Off. J. Soc. Neurosci. 22 (2002) 6019e6028.
[15] A.L. Kolpak, J. Jiang, D. Guo, C. Standley, K. Bellve, K. Fogarty, Z.Z. Bao,
Negative guidance factor-induced macropinocytosis in the growth cone
plays a critical role in repulsive axon turning, J. Neurosci. Off. J. Soc. Neurosci.
29 (2009) 10488e10498.
[16] J.H. Jang, C.S. Lee, D. Hwang, S.H. Ryu, Understanding of the roles of phospholipase D and phosphatidic acid through their binding partners, Prog.
Lipid Res. 51 (2012) 71e81.
[17] Y. Liu, Y. Su, X. Wang, Phosphatidic acid-mediated signaling, Adv. Exp. Med.
Biol. 991 (2013) 159e176.
[18] C.L. Stace, N.T. Ktistakis, Phosphatidic acid- and phosphatidylserine-binding
proteins, Biochim. Biophys. Acta 1761 (2006) 913e926.
[19] J.E. Vance, D.E. Vance, Phospholipid biosynthesis in mammalian cells, Biochem. Cell. Biol. 82 (2004) 113e128.
[20] D.E. Vance, H. Goldne, Konrad Blochea pioneer in cholesterol and fatty acid
biosynthesis, Biochem. Biophys. Res. Commun. 292 (2002) 1117e1127.
[21] A.J. Morris, Regulation of phospholipase D activity, membrane targeting and
intracellular trafcking by phosphoinositides, Biochem. Soc. Symp. (2007)
247e257.
[22] V. Chalifa-Caspi, Y. Eli, M. Liscovitch, Kinetic analysis in mixed micelles of
partially puried rat brain phospholipase D activity and its activation by
phosphatidylinositol 4,5-bisphosphate, Neurochem. Res. 23 (1998) 589e599.
[23] M.A. Frohman, A.J. Morris, Phospholipase D structure and regulation, Chem.
Phys. Lipids 98 (1999) 127e140.
[24] T.C. Sung, Y. Zhang, A.J. Morris, M.A. Frohman, Structural analysis of human
phospholipase D1, J. Biol. Chem. 274 (1999) 3659e3666.
[25] T.C. Sung, Y.M. Altshuller, A.J. Morris, M.A. Frohman, Molecular analysis of
mammalian phospholipase D2, J. Biol. Chem. 274 (1999) 494e502.
[26] M. Hiroyama, J.H. Exton, Studies of the roles of ADP-ribosylation factors and
phospholipase D in phorbol ester-induced membrane rufing, J. Cell. Phys.
202 (2005) 608e622.
[27] M. Rankovic, L. Jacob, V. Rankovic, L.O. Brandenburg, H. Schroder, V. Hollt,
T. Koch, ADP-ribosylation factor 6 regulates mu-opioid receptor trafcking
and signaling via activation of phospholipase D2, Cell. Signal. 21 (2009)
1784e1793.
[28] H.J. Peng, K.M. Henkels, M. Mahankali, C. Marchal, P. Bubulya, M.C. Dinauer,
J. Gomez-Cambronero, The dual effect of Rac2 on phospholipase D2 regulation that explains both the onset and termination of chemotaxis, Mol. Cell.
Biol. 31 (2011) 2227e2240.
[29] M. Osisami, W. Ali, M.A. Frohman, A role for phospholipase D3 in myotube
formation, PloS One 7 (2012) e33341.
[30] Y. Otani, Y. Yamaguchi, Y. Sato, T. Furuichi, K. Ikenaka, H. Kitani, H. Baba,
PLD4 is involved in phagocytosis of microglia: expression and localization
changes of PLD4 are correlated with activation state of microglia, PloS One 6
(2011) e27544.
[31] M.K. Topham, Signaling roles of diacylglycerol kinases, J. Cell. Biochem. 97
(2006) 474e484.
[32] G.M. Jenkins, M.A. Frohman, Phospholipase D: a lipid centric review, Cell.
Mol. Life Sci. CMLS 62 (2005) 2305e2316.
[33] W. Haque, A. Garg, A.K. Agarwal, Enzymatic activity of naturally occurring 1acylglycerol-3-phosphate-O-acyltransferase 2 mutants associated with
congenital generalized lipodystrophy, Biochem. Biophys. Res. Commun. 327
(2005) 446e453.
[34] D.W. Leung, The structure and functions of human lysophosphatidic acid
acyltransferases, Front. Biosci. J. Virtual Libr. 6 (2001) D944eD953.
[35] T.M. Lewin, P. Wang, R.A. Coleman, Analysis of amino acid motifs diagnostic
for the sn-glycerol-3-phosphate acyltransferase reaction, Biochemistry 38
(1999) 5764e5771.
[36] L.K. Dircks, J. Ke, H.S. Sul, A conserved seven amino acid stretch important
for murine mitochondrial glycerol-3-phosphate acyltransferase activity.

56

[37]

[38]
[39]

[40]

[41]

[42]
[43]

[44]

[45]

[46]

[47]

[48]

[49]

[50]

[51]
[52]

[53]
[54]
[55]

[56]

[57]

[58]

[59]

[60]

[61]

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57


Signicance of arginine 318 in catalysis, J. Biol. Chem. 274 (1999)
34728e34734.
K. Schwarz, S. Natarajan, N. Kassas, N. Vitale, F. Schmitz, The synaptic ribbon
is a site of phosphatidic acid generation in ribbon synapses, J. Neurosci. Off. J.
Soc. Neurosci. 31 (2011) 15996e16011.
A.E. Traynor, D. Schubert, W.R. Allen, Alterations of lipid metabolism in
response to nerve growth factor, J. Neurochem. 39 (1982) 1677e1683.
Y. Kanaho, Y. Funakoshi, H. Hasegawa, Phospholipase D signalling and its
involvement in neurite outgrowth, Biochim. Biophys. Acta 1791 (2009)
898e904.
Y. Hozumi, M. Watanabe, K. Otani, K. Goto, Diacylglycerol kinase beta promotes dendritic outgrowth and spine maturation in developing hippocampal
neurons, BMC Neurosci. 10 (2009) 99.
Y. Shirai, T. Kouzuki, K. Kakefuda, S. Moriguchi, A. Oyagi, K. Horie, S.Y. Morita,
M. Shimazawa, K. Fukunaga, J. Takeda, N. Saito, H. Hara, Essential role of
neuron-enriched diacylglycerol kinase (DGK), DGKbeta in neurite spine
formation, contributing to cognitive function, PloS One 5 (2010) e11602.
E.E. Govek, S.E. Newey, L. Van Aelst, The role of the Rho GTPases in neuronal
development, Genes. Dev. 19 (2005) 1e49.
A.J. Ridley, A. Hall, The small GTP-binding protein rho regulates the assembly
of focal adhesions and actin stress bers in response to growth factors, Cell
70 (1992) 389e399.
Y.C. Chae, J.H. Kim, K.L. Kim, H.W. Kim, H.Y. Lee, W.D. Heo, T. Meyer, P.G. Suh,
S.H. Ryu, Phospholipase D activity regulates integrin-mediated cell spreading
and migration by inducing GTP-Rac translocation to the plasma membrane,
Mol. Biol. Cell. 19 (2008) 3111e3123.
D. Faugaret, F.C. Chouinard, D. Harbour, M.A. El azreq, S.G. Bourgoin, An
essential role for phospholipase D in the recruitment of vesicle amine
transport protein-1 to membranes in human neutrophils, Biochem. Pharmacol. 81 (2011) 144e156.
F. Sanematsu, A. Nishikimi, M. Watanabe, T. Hongu, Y. Tanaka, Y. Kanaho,
J.F. Cote, Y. Fukui, Phosphatidic acid-dependent recruitment and function of
the Rac activator DOCK1 during dorsal rufe formation, J. Biol. Chem. 288
(2013) 8092e8100.
A. Nishikimi, H. Fukuhara, W. Su, T. Hongu, S. Takasuga, H. Mihara, Q. Cao,
F. Sanematsu, M. Kanai, H. Hasegawa, Y. Tanaka, M. Shibasaki, Y. Kanaho,
T. Sasaki, M.A. Frohman, Y. Fukui, Sequential regulation of DOCK2 dynamics
by two phospholipids during neutrophil chemotaxis, Science 324 (2009)
384e387.
T. Kurooka, Y. Yamamoto, Y. Takai, T. Sakisaka, Dual regulation of RA-RhoGAP
activity by phosphatidic acid and Rap1 during neurite outgrowth, J. Biol.
Chem. 286 (2011) 6832e6843.
H. Abramovici, P. Mojtabaie, R.J. Parks, X.P. Zhong, G.A. Koretzky,
M.K. Topham, S.H. Gee, Diacylglycerol kinase zeta regulates actin cytoskeleton reorganization through dissociation of Rac1 from RhoGDI, Mol. Biol.
Cell. 20 (2009) 2049e2059.
R.H. Daniels, P.S. Hall, G.M. Bokoch, Membrane targeting of p21-activated
kinase 1 (PAK1) induces neurite outgrowth from PC12 cells, EMBO J. 17
(1998) 754e764.
K. Hayashi, T. Ohshima, M. Hashimoto, K. Mikoshiba, Pak1 regulates dendritic branching and spine formation, Dev. Neurobiol. 67 (2007) 655e669.
P. Penzes, A. Beeser, J. Chernoff, M.R. Schiller, B.A. Eipper, R.E. Mains,
R.L. Huganir, Rapid induction of dendritic spine morphogenesis by transsynaptic ephrinB-EphB receptor activation of the Rho-GEF kalirin, Neuron
37 (2003) 263e274.
L.C. Sanders, F. Matsumura, G.M. Bokoch, P. de Lanerolle, Inhibition of myosin
light chain kinase by p21-activated kinase, Science 283 (1999) 2083e2085.
P.D. Sarmiere, J.R. Bamburg, Regulation of the neuronal actin cytoskeleton by
ADF/colin, J. Neurobiol. 58 (2004) 103e117.
R.K. Vadlamudi, F. Li, C.J. Barnes, R. Bagheri-Yarmand, R. Kumar, p41-Arc
subunit of human Arp2/3 complex is a p21-activated kinase-1-interacting
substrate, EMBO Rep. 5 (2004) 154e160.
N. Divecha, M. Roefs, J.R. Halstead, S. D'Andrea, M. Fernandez-Borga,
L. Oomen, K.M. Saqib, M.J. Wakelam, C. D'Santos, Interaction of the type
Ialpha PIPkinase with phospholipase D: a role for the local generation of
phosphatidylinositol 4, 5-bisphosphate in the regulation of PLD2 activity,
EMBO J. 19 (2000) 5440e5449.
M. Jarquin-Pardo, A. Fitzpatrick, F.J. Galiano, E.A. First, J.N. Davis, Phosphatidic acid regulates the afnity of the murine phosphatidylinositol 4phosphate 5-kinase-Ibeta for phosphatidylinositol-4-phosphate, J. Cell. Biochem. 100 (2007) 112e128.
A.N. Roach, Z. Wang, P. Wu, F. Zhang, R.B. Chan, Y. Yonekubo, G. Di Paolo,
A.A. Gorfe, G. Du, Phosphatidic acid regulation of PIPKI is critical for actin
cytoskeletal reorganization, J. Lipid Res. 53 (2012) 2598e2609.
A. Honda, M. Nogami, T. Yokozeki, M. Yamazaki, H. Nakamura, H. Watanabe,
K. Kawamoto, K. Nakayama, A.J. Morris, M.A. Frohman, Y. Kanaho, Phosphatidylinositol 4-phosphate 5-kinase alpha is a downstream effector of the
small G protein ARF6 in membrane rufe formation, Cell 99 (1999) 521e532.
B. Luo, S.M. Prescott, M.K. Topham, Diacylglycerol kinase zeta regulates
phosphatidylinositol 4-phosphate 5-kinase Ialpha by a novel mechanism,
Cell. Signal. 16 (2004) 891e897.
H. Ishihara, Y. Shibasaki, N. Kizuki, T. Wada, Y. Yazaki, T. Asano, Y. Oka, Type I
phosphatidylinositol-4-phosphate 5-kinases. Cloning of the third isoform
and deletion/substitution analysis of members of this novel lipid kinase
family, J. Biol. Chem. 273 (1998) 8741e8748.

[62] I. van den Bout, N. Divecha, PIP5K-driven PtdIns(4,5)P2 synthesis: regulation


and cellular functions, J. Cell. Sci. 122 (2009) 3837e3850.
[63] M. Yamazaki, Y. Yamauchi, Y. Goshima, Y. Kanaho, Phosphatidylinositol 4phosphate 5-kinase beta regulates growth cone morphology and semaphorin 3A-triggered growth cone collapse in mouse dorsal root ganglion
neurons, Neurosci. Lett. 547 (2013) 59e64.
[64] M.L. Giudici, K.A. Hinchliffe, R.F. Irvine, Phosphatidylinositol phosphate kinases, J. Endocrinol. Invest. 27 (2004) 137e142.
[65] D.J. Hernandez-Deviez, M.G. Roth, J.E. Casanova, J.M. Wilson, ARNO and ARF6
regulate axonal elongation and branching through downstream activation of
phosphatidylinositol 4-phosphate 5-kinase alpha, Mol. Biol. Cell. 15 (2004)
111e120.
[66] S. Corbalan-Garcia, S. Sanchez-Carrillo, J. Garcia-Garcia, J.C. Gomez-Fernandez, Characterization of the membrane binding mode of the C2 domain of
PKC epsilon, Biochemistry 42 (2003) 11661e11668.
[67] M. Jose Lopez-Andreo, J.C. Gomez-Fernandez, S. Corbalan-Garcia, The
simultaneous production of phosphatidic acid and diacylglycerol is essential
for the translocation of protein kinase Cepsilon to the plasma membrane in
RBL-2H3 cells, Mol. Biol. Cell. 14 (2003) 4885e4895.
[68] T. Yokozeki, K. Homma, S. Kuroda, U. Kikkawa, S. Ohno, M. Takahashi,
K. Imahori, Y. Kanaho, Phosphatidic acid-dependent phosphorylation of a 29kDa protein by protein kinase Calpha in bovine brain cytosol, J. Neurochem.
71 (1998) 410e417.
[69] C. Larsson, Protein kinase C and the regulation of the actin cytoskeleton, Cell.
Signal. 18 (2006) 276e284.
[70] S. Ito, N. Richert, I. Pastan, Phospholipids stimulate phosphorylation of vinculin by the tyrosine-specic protein kinase of Rous sarcoma virus, Proc.
Natl. Acad. Sci. U S A 79 (1982) 4628e4631.
[71] R.P. Johnson, S.W. Craig, F-actin binding site masked by the intramolecular
association of vinculin head and tail domains, Nature 373 (1995)
261e264.
[72] M.F. Bader, N. Vitale, Phospholipase D in calcium-regulated exocytosis: lessons from chromafn cells, Biochim. Biophys. Acta 1791 (2009) 936e941.
[73] M.R. Ammar, N. Kassas, S. Chasserot-Golaz, M.F. Bader, N. Vitale, Lipids in
regulated exocytosis: what are they doing? Front. Endocrinol. 4 (2013) 125.
[74] N. Vitale, A.S. Caumont, S. Chasserot-Golaz, G. Du, S. Wu, V.A. Sciorra,
A.J. Morris, M.A. Frohman, M.F. Bader, Phospholipase D1: a key factor for the
exocytotic machinery in neuroendocrine cells, EMBO J. 20 (2001)
2424e2434.
[75] M. Zeniou-Meyer, N. Zabari, U. Ashery, S. Chasserot-Golaz, A.M. Haeberle,
V. Demais, Y. Bailly, I. Gottfried, H. Nakanishi, A.M. Neiman, G. Du,
M.A. Frohman, M.F. Bader, N. Vitale, Phospholipase D1 production of phosphatidic acid at the plasma membrane promotes exocytosis of large densecore granules at a late stage, J. Biol. Chem. 282 (2007) 21746e21757.
[76] A.D. Lam, P. Tryoen-Toth, B. Tsai, N. Vitale, E.L. Stuenkel, SNARE-catalyzed
fusion events are regulated by Syntaxin1A-lipid interactions, Mol. Biol. Cell.
19 (2008) 485e497.
[77] M.R. Ammar, Y. Humeau, A. Hanauer, B. Nieswandt, M.F. Bader, N. Vitale, The
Cofn-Lowry syndrome-associated protein RSK2 regulates neurite
outgrowth through phosphorylation of phospholipase D1 (PLD1) and synthesis of phosphatidic acid, J. Neurosci. Off. J. Soc. Neurosci. 33 (2013)
19470e19479.
[78] G. Lalli, A. Hall, Ral GTPases regulate neurite branching through GAP-43 and
the exocyst complex, J. Cell. Biol. 171 (2005) 857e869.
[79] Y.B. Zhu, K. Kang, Y. Zhang, C. Qi, G. Li, D.M. Yin, Y. Wang, PLD1 negatively
regulates dendritic branching, J. Neurosci. Off. J. Soc. Neurosci. 32 (2012)
7960e7969.
[80] K. Kim, J. Yang, X.P. Zhong, M.H. Kim, Y.S. Kim, H.W. Lee, S. Han, J. Choi,
K. Han, J. Seo, S.M. Prescott, M.K. Topham, Y.C. Bae, G. Koretzky, S.Y. Choi,
E. Kim, Synaptic removal of diacylglycerol by DGKzeta and PSD-95 regulates
dendritic spine maintenance, EMBO J. 28 (2009) 1170e1179.
[81] K. Kim, J. Yang, E. Kim, Diacylglycerol kinases in the regulation of dendritic
spines, J. Neurochem. 112 (2010) 577e587.
[82] H. Zhang, D.J. Webb, H. Asmussen, S. Niu, A.F. Horwitz, A GIT1/PIX/Rac/PAK
signaling module regulates spine morphogenesis and synapse formation
through MLC, J. Neurosci. Off. J. Soc. Neurosci. 25 (2005) 3379e3388.
[83] B. Boda, I. Nikonenko, S. Alberi, D. Muller, Central nervous system functions
of PAK protein family: from spine morphogenesis to mental retardation, Mol.
Neurobiol. 34 (2006) 67e80.
[84] K.A. Jones, D.P. Srivastava, J.A. Allen, R.T. Strachan, B.L. Roth, P. Penzes, Rapid
modulation of spine morphology by the 5-HT2A serotonin receptor through
kalirin-7 signaling, Proc. Natl. Acad. Sci. U S A 106 (2009) 19575e19580.
[85] M. Ackermann, A. Matus, Activity-induced targeting of prolin and stabilization of dendritic spine morphology, Nat. Neurosci. 6 (2003) 1194e1200.
[86] T. Nakagawa, J.A. Engler, M. Sheng, The dynamic turnover and functional
roles of alpha-actinin in dendritic spines, Neuropharmacology 47 (2004)
734e745.
[87] J. Choi, J. Ko, B. Racz, A. Burette, J.R. Lee, S. Kim, M. Na, H.W. Lee, K. Kim,
R.J. Weinberg, E. Kim, Regulation of dendritic spine morphogenesis by insulin receptor substrate 53, a downstream effector of Rac1 and Cdc42 small
GTPases, J. Neurosci. Off. J. Soc. Neurosci. 25 (2005) 869e879.
[88] Y. Kim, J.Y. Sung, I. Ceglia, K.W. Lee, J.H. Ahn, J.M. Halford, A.M. Kim,
S.P. Kwak, J.B. Park, S. Ho Ryu, A. Schenck, B. Bardoni, J.D. Scott, A.C. Nairn,
P. Greengard, Phosphorylation of WAVE1 regulates actin polymerization and
dendritic spine morphology, Nature 442 (2006) 814e817.

M.-R. Ammar et al. / Biochimie 107 (2014) 51e57


[89] A.M. Wegner, C.A. Nebhan, L. Hu, D. Majumdar, K.M. Meier, A.M. Weaver,
D.J. Webb, N-wasp and the arp2/3 complex are critical regulators of actin in
the development of dendritic spines and synapses, J. Biol. Chem. 283 (2008)
15912e15920.
[90] C. Stace, M. Manifava, C. Delon, J. Coadwell, S. Cockcroft, N.T. Ktistakis, PA
binding of phosphatidylinositol 4-phosphate 5-kinase, Adv. Enzyme Regul.
48 (2008) 55e72.
[91] K.F. Tolias, A.D. Couvillon, L.C. Cantley, C.L. Carpenter, Characterization of a
Rac1- and RhoGDI-associated lipid kinase signaling complex, Mol. Cell. Biol.
18 (1998) 762e770.
[92] H. Zhang, I.G. Macara, The PAR-6 polarity protein regulates dendritic spine
morphogenesis through p190 RhoGAP and the Rho GTPase, Dev. Cell. 14
(2008) 216e226.
[93] J.A. Jones, R. Rawles, Y.A. Hannun, Identication of a novel phosphatidic acid
binding domain in protein phosphatase-1, Biochemistry 44 (2005)
13235e13245.
[94] S. Strack, S. Kini, F.F. Ebner, B.E. Wadzinski, R.J. Colbran, Differential cellular
and subcellular localization of protein phosphatase 1 isoforms in brain,
J. Comp. Neurol. 413 (1999) 373e384.
[95] J.D. Richter, E. Klann, Making synaptic plasticity and memory last: mechanisms of translational regulation, Genes. Dev. 23 (2009) 1e11.
[96] R.T. Terry-Lorenzo, D.W. Roadcap, T. Otsuka, T.A. Blanpied, P.L. Zamorano,
C.C. Garner, S. Shenolikar, M.D. Ehlers, Neurabin/protein phosphatase-1

[97]

[98]

[99]

[100]

[101]

[102]

57

complex regulates dendritic spine morphogenesis and maturation, Mol.


Biol. Cell. 16 (2005) 2349e2362.
J. Jaworski, S. Spangler, D.P. Seeburg, C.C. Hoogenraad, M. Sheng, Control of
dendritic arborization by the phosphoinositide-30 -kinase-Akt-mammalian
target of rapamycin pathway, J. Neurosci. Off. J. Soc. Neurosci. 25 (2005)
11300e11312.
V. Kumar, M.X. Zhang, M.W. Swank, J. Kunz, G.Y. Wu, Regulation of dendritic
morphogenesis by Ras-PI3K-Akt-mTOR and Ras-MAPK signaling pathways,
J. Neurosci. Off. J. Soc. Neurosci. 25 (2005) 11288e11299.
N. Kassas, P. Tryoen-Toth, M. Corrotte, T. Thahouly, M.F. Bader, N.J. Grant,
N. Vitale, Genetically encoded probes for phosphatidic acid, Methods Cell.
Biol. 108 (2012) 445e459.
R.A. Haast, A.J. Kiliaan, Impact of fatty acids on brain circulation, structure
and function, Prostagl. Leukot. Essent. Fat. Acids (2014), http://dx.doi.org/
10.1016/j.plefa.2014.01.002 [Epub ahead of print].
U. Burkhardt, D. Stegner, E. Hattingen, S. Beyer, J. B Nieswandt, Klein
impaired brain development and reduced cognitive function in phospholipase D-decient mice, Neurosci. Lett. (2014) 48e52.
T.G. Oliveira, R.B. Chan, H. Tian, M. Laredo, G. Shui, A. Staniszewski, H. Zhang,
L. Wang, T.W. Kim, K.E. Duff, M.R. Wenk, O. Arancio, G. Di Paolo, Phospholipase d2 ablation ameliorates Alzheimer's disease-linked synaptic
dysfunction and cognitive decits, J. Neurosci. (2010) 16419e16428.

You might also like