ArticlePDF Available

Improved cognition, mild anxiety-like behavior and decreased motor performance in pyridoxal phosphatase-deficient mice

Authors:

Abstract and Figures

Pyridoxal 5′-phosphate (PLP) is an essential cofactor in the catalysis of ~140 different enzymatic reactions. A pharmacological elevation of cellular PLP concentrations is of interest in neuropsychiatric diseases, but whole-body consequences of higher intracellular PLP levels are unknown. To address this question, we have generated mice allowing a conditional ablation of the PLP phosphatase PDXP. Ubiquitous PDXP deletion increased PLP levels in brain, skeletal muscle and red blood cells up to 3-fold compared to control mice, demonstrating that PDXP acts as a major regulator of cellular PLP concentrations in vivo. Neurotransmitter analysis revealed that the concentrations of dopamine, serotonin, epinephrine and glutamate were unchanged in the brains of PDXP knockout mice. However, the levels of γ-aminobutyric acid (GABA) increased by ~20%, demonstrating that elevated PLP levels can drive additional GABA production. Behavioral phenotyping of PDXP knockout mice revealed improved spatial learning and memory, and a mild anxiety-like behavior. Consistent with elevated GABA levels in the brain, PDXP loss in neural cells decreased performance in motor tests, whereas PDXP-deficiency in skeletal muscle increased grip strength. Our findings suggest that PDXP is involved in the fine-tuning of GABA biosynthesis. Pharmacological inhibition of PDXP might correct the excitatory/inhibitory imbalance in some neuropsychiatric diseases.
Content may be subject to copyright.
Contents lists available at ScienceDirect
BBA - Molecular Basis of Disease
journal homepage: www.elsevier.com/locate/bbadis
Improved cognition, mild anxiety-like behavior and decreased motor
performance in pyridoxal phosphatase-decient mice
Elisabeth Jeanclos
a,
, Monique Albersen
b,1
, Rúben J.J. Ramos
b
, Annette Raab
a,c
,
Christian Wilhelm
a
, Leif Hommers
a,c,d
, Klaus-Peter Lesch
c,d,e,f,g
, Nanda M. Verhoeven-Duif
b
,
Antje Gohla
a,
a
Institute of Pharmacology and Toxicology, University of Würzburg, Germany
b
Department of Genetics, University Medical Center Utrecht, the Netherlands
c
Interdisciplinary Center for Clinical Research, University Hospital Würzburg, Germany
d
Comprehensive Heart Failure Center, University Hospital Würzburg, Germany
e
Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany
f
Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
g
Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
ARTICLE INFO
Keywords:
Pyridoxal phosphatase
Vitamin B6
γ-Aminobutyric acid (GABA)
Motor performance
Neuropsychiatric diseases
Neurotransmitter biosynthesis
ABSTRACT
Pyridoxal 5-phosphate (PLP) is an essential cofactor in the catalysis of ~140 dierent enzymatic reactions. A
pharmacological elevation of cellular PLP concentrations is of interest in neuropsychiatric diseases, but whole-
body consequences of higher intracellular PLP levels are unknown. To address this question, we have generated
mice allowing a conditional ablation of the PLP phosphatase PDXP. Ubiquitous PDXP deletion increased PLP
levels in brain, skeletal muscle and red blood cells up to 3-fold compared to control mice, demonstrating that
PDXP acts as a major regulator of cellular PLP concentrations in vivo. Neurotransmitter analysis revealed that the
concentrations of dopamine, serotonin, epinephrine and glutamate were unchanged in the brains of PDXP
knockout mice. However, the levels of γ-aminobutyric acid (GABA) increased by ~20%, demonstrating that
elevated PLP levels can drive additional GABA production. Behavioral phenotyping of PDXP knockout mice
revealed improved spatial learning and memory, and a mild anxiety-like behavior. Consistent with elevated
GABA levels in the brain, PDXP loss in neural cells decreased performance in motor tests, whereas PDXP-de-
ciency in skeletal muscle increased grip strength. Our ndings suggest that PDXP is involved in the ne-tuning of
GABA biosynthesis. Pharmacological inhibition of PDXP might correct the excitatory/inhibitory imbalance in
some neuropsychiatric diseases.
1. Introduction
Pyridoxal 5-phosphate (PLP), the co-enzymatically active form of
vitamin B6, is one of the most versatile cofactors found in nature. In
mammals, PLP-dependent enzymes catalyze ~140 dierent types of
biochemical transformations, including reactions that are required for
neurotransmitter-, amino acid- and glycogen metabolism [1]. PLP is a
highly reactive aldehyde that transiently forms a Schibase with the ε-
amino group of the active site lysine in its cognate apo-enzymes [2]. It
is thought that intracellular PLP concentrations and PLP tracking are
tightly controlled to ensure sucient PLP supply to apo-enzymes, while
minimizing free PLP levels. This is important to prevent non-specic
reactions of PLP with cellular nucleophiles and proteins that are not B6
enzymes [35].
Intracellular PLP levels depend on the availability and -transport of
PLP precursors [6], the biosynthetic activities of pyridoxal 5-kinase
(PDXK) and pyridox(am)ine 5-phosphate oxidase (PNPO), the extent of
PLP scavenging by proteins and small molecules, PLP binding to carriers
such as PROSC, PDXK and PNPO, and PLP hydrolysis by pyridoxal 5-
phosphate phosphatase (PDXP) [3,4,7]. Specically, PDXK catalyzes the
phosphorylation of pyridoxine (PN), pyridoxamine (PM) or pyridoxal (PL)
to their 5-phosphorylated variants PNP, PMP or PLP. PNPO additionally
(re)generates PLP from PNP or PMP by catalyzing the oxidation of their
respective hydroxyl or amino groups to the co-enzymatically essential
https://doi.org/10.1016/j.bbadis.2018.08.018
Received 29 June 2018; Received in revised form 13 August 2018; Accepted 14 August 2018
Corresponding authors.
1
Current address: Department of Clinical Chemistry, VU Medical Center, Amsterdam, the Netherlands.
E-mail addresses: elisabeth.jeanclos@uni-wuerzburg.de (E. Jeanclos), antje.gohla@uni-wuerzburg.de (A. Gohla).
BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
0925-4439/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
Please cite this article as: Jeanclos, E., BBA - Molecular Basis of Disease (2018), https://doi.org/10.1016/j.bbadis.2018.08.018
aldehyde moiety of PLP [3](Fig. 1). The pathways of PLP biosynthesis
have been established for decades, yet a dedicated PLP phosphatase
(PDXP) was cloned more recently [8]. PDXP activity may play a signicant
role in the regulation of cellular PLP concentrations [7,8]. Nonetheless, the
relative importance of PDXP for the regulation of PLP homeostasis and for
PLP-dependent functions in vivo is unknown.
A pharmacological elevation of cellular PLP concentrations is of
interest in neuropsychiatric disorders and inammation [911]. Inborn
errors of vitamin B6 metabolism can lead to inadequate levels of PLP in
the brain and cause childhood epilepsy [4,1217]. In these cases, the
administration of PLP or PN leads to prompt seizure cessation. How-
ever, high doses of PN can cause respiratory depression, and impose
treatment withdrawal [4]. Individuals with B6-dependent epilepsy
often require continuous oral PN supplementation, yet long-term
treatment with high doses of PN or PLP can be toxic [1820]. A phar-
macological inhibition of PDXP [21] might circumvent toxic eects
caused by elevated extracellular B6 vitamer levels, and provide a novel
approach to increase cellular PLP concentrations.
The aim of the present study was to analyze the relative importance
of PDXP for the regulation of tissue PLP concentrations, and to explore
the biochemical and behavioral consequences of PDXP deletion in gene-
targeted mice. Our results demonstrate that PDXP is a major factor in
the control of cellular PLP concentrations in vivo, and we uncover a
previously unexploited approach to increase GABA levels in the brain.
PDXP loss improved spatial learning and memory, but also caused a
mild anxiety-like phenotype and central nervous system-dependent
muscle weakness. Further research is necessary to evaluate whether
pharmacological PDXP inhibition might be an advantageous strategy to
increase cellular PLP levels.
2. Materials and methods
2.1. Materials
Unless otherwise specied, all reagents were of the highest available
purity and purchased from Sigma-Aldrich.
2.2. Generation and breeding of Pdxp knockout mice
Floxed Pdxp mice (Pdxp
tm1Goh
) were generated on a C57Bl/6J
background by Ozgene Pty Ltd., Australia. The neomycin resistance
cassette was removed by breeding with the global FLPe deleter strain
B6.129S4-Gt(ROSA)26Sor < tm1(FLP1)Dym > /RainJ. Whole-body,
skeletal muscle or neural cell-directed Pdxp knockouts were achieved
by breeding with B6.FVB-Tg(EIIa-cre)C5379Lmgd/J (EIIa-Cre), B6.Cg-
Tg(ACTA1-cre)79Jme/J (Acta1-Cre), or B6.Cg-Tg(Nes-cre)1Kln/J
(Nestin-Cre) transgenic mice obtained from The Jackson Laboratory.
Mouse experiments were approved by the local authorities (Regierung
Unterfranken), and all analyses were carried out in strict accordance
with all German and European Union applicable laws and regulations
concerning care and use of laboratory animals.
2.3. Genotyping
Genomic DNA was isolated with DNeasy (Qiagen) and digested with
EcoRV. Southern blotting was performed according to standard proce-
dures. Briey, DNA was subjected to agarose gel electrophoresis, de-
natured, blotted onto nitrocellulose and crosslinked with UV light. The
probe was generated by PCR using Pfx polymerase (Invitrogen) and
labeled with [α-
32
P] dCTP (Hartmann Analytic), employing the
DecaPrime II DNA labeling kit (Ambion). Unincorporated labeled nu-
cleotides were removed with Illustra ProbeQuant G-50 micro columns
(GE Healthcare). Genotyping of Acta1-Cre and Nestin-Cre transgenic
mice was performed by PCR as described by The Jackson Laboratory.
2.4. Tissue sample preparation
Mice were sacriced by cervical dislocation and perfused with PBS.
Skeletal muscle samples were taken from the upper back hind limb, and
contained musculus biceps femoris,musculus semimembranosus and mus-
culus semitendinosus. Small intestine samples (comprising duodenum,
jejunum and ileum) were cut into small segments and washed ex-
tensively with phosphate-buered saline (PBS). For liver analysis, the
entire organ was used. Brains were either used in toto,ordierent brain
regions were dissected. To this end, the brain was placed on an ice-cold
adult mouse brain slicer (Harvard Apparatus). A 3 mm coronal section
was cut starting from the basis of the circle of Willis towards the frontal
part of the brain (Bregma 3.16 to Bregma 0.02), and the brain cortex
and the midbrain region (containing hippocampus, hypothalamus and
thalamus) were dissected from this slice. All brain parts including cer-
ebellum and hindbrain were weighed (wet weight). Pups were sacri-
ced by decapitation on postnatal day 6 (P6), and brains were
PL PMPN
PLP PMPPNP
HH H
PO32-
PNPO PNPO
PNPO PNPO
PDXK
PDXP PDXK
PDXP
PO32- PO32-
PA
AOX DH
PDXK
PDXP
Fig. 1. Enzymes involved in mammalian
PLP metabolism. PA, 4-pyridoxic acid;
PN(P), pyridoxine (-5-phosphate); PL(P),
pyridoxal (-5-phosphate); PM(P), pyr-
idoxamine (-5-phosphate). PDXP, pyr-
idoxal phosphatase; PDXK, pyridoxal
kinase; PNPO, pyridox(am)ine oxidase;
AOX/DH, aldehyde oxidase/β-NAD de-
hydrogenase. PL is oxidized to PA in the
liver and excreted into the urine.
Catalytic eciencies (k
cat
/K
M
) of PDXP:
PLP, 6.1 × 10
5
M
1
×s
1
; PNP, 2.7 ×
10
4
M
1
×s
1
; PMP, 5.6 × 10
3
M
1
×s
1
[8].
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
2
dissected, weighed and ash-frozen in liquid nitrogen. For analysis,
organs and tissue parts were homogenized in ice-cold PBS with a Potter
S homogenizer (Sartorius). Proteins were precipitated with 10% (w/v)
6.1-N trichloroacetic acid (TCA) for 15 min on ice with occasional
vortexing. After centrifugation (20,000 ×g, 15 min, 4 °C), supernatants
were ash-frozen in liquid nitrogen and kept at 80 °C.
2.5. Plasma and red blood cell preparation
Heparinized blood was centrifuged at room temperature (800 ×g,
10 min). The supernatant (plasma) was centrifuged again, and proteins
in the supernatant were removed by precipitation, using 10% (w/v)
6.1-N TCA. Packed red blood cells (RBCs) were washed by centrifuga-
tion and resuspension in 0.9% (w/v) NaCl. Cells were lysed and pro-
teins were precipitated by adding 10% 6.1-N TCA. Plasma and RBC
extracts were ash-frozen in liquid nitrogen and kept at 80 °C until
analysis. To calculate the intracellular concentrations of B
6
vitamers,
the intracellular RBC volume was estimated to correspond to 52% of the
packed RBC volume.
2.6. Quantication of B
6
vitamers, GABA and amino acids
B
6
vitamers, GABA and other amino acids were quantied using
UPLC-MS/MS as described [6,2224].
2.7. Neurotransmitter analysis by HPLC
Mice were sacriced by cervical dislocation, brains were dissected,
weighed and ash-frozen in liquid nitrogen. The entire procedure was
performed in < 3 min. Frozen brains were sonicated (2 × 30 s) in a
0.3 M perchloric acid solution at 4 °C, and samples were centrifuged
(16,000 ×g, 10 min, 4 °C). Supernatants were analyzed on a Dionex
Ultimate 3000 HPLC (ThermoScientic), using 75 mM NaH
2
PO
4
,
1.5 mM octane sulfonic acid, 10% (v/v) acetonitrile; pH 3 as a mobile
phase. Neurotransmitters were separated on a 3 μm reverse phase
column (BDS-HYPERSIL-C18, ThermoScientic) and detected by elec-
trochemical reaction at 450 mV.
2.8. Western blot analysis
Tissue or cell homogenates were prepared as detailed above and
extracted with RIPA buer [50 mM Tris, pH 7.5; 150mM NaCl, 1% (v/v)
Triton X-100, 0.5% (v/v) sodium deoxycholate, 0.1% (w/v) SDS, 1 mM
4-(2-aminoethyl)benzenesulfonyl uoride (Pefabloc), 5 μg/mL aprotinin,
1μg/mL leupeptin, 1 μg/mL pepstatin] for 15 min at 4 °C under rotation,
and lysates were claried by centrifugation (20,000 ×g, 15 min, 4 °C).
Protein concentrations in the supernatants were determined using the
Micro BCA Protein Assay Kit (Thermo Scientic). Samples were solubi-
lized in Laemmli buer and subjected to standard immunoblotting. The
following antibodies were used: Cell Signaling Technology, α-PDXP
(clone C85E3) and α-GAPDH (clone 14C10); Merck/Millipore, α-actin
(mAb1501) and α-FAK (clone 4.47); ThermoFisher Scientic, α-PNPO
(#PA5-26400); and Sigma Aldrich, α-PDXK (AB1, #AV53615). To ana-
lyze Ser3-phosphocolin and colin levels, tissues were dissected from 4-
months old male mice, and immediately ash-frozen in liquid nitrogen as
described above. Frozen tissues were then pulverized using a pre-cooled
porcelain mortar and pestle. About 50100 mg of this powder was so-
lubilized in 250 μLice-coldlysisbuer [20 mM Tris, pH 7.5; 150 mM
NaCl, 1% (v/v) Triton X-100, 1 mM β-glycerophosphate, 2.5 mM sodium
pyrophosphate, 1 mM sodium orthovanadate, 1 mM Pefabloc, 1 μg/mL
leupeptin/aprotinin/pepstatin; 1/1000 phosphatase inhibitor cocktail I
and II (both from Sigma Aldrich, #P2850 and P5726)] for 2 h at 4 °C
under rotation. Lysates were cleared by centrifugation (20,000 ×g,
15 min, 4 °C), and protein concentrations in the supernatants were de-
termined and adjusted to ~20 μg/μL using the Micro BCA Protein Assay
Kit. Samples were diluted to a nal concentration of ~2 μg/μLin2×
Laemmli buer, and ~4050 μgofproteinswereseparatedona12%
SDS-PAGE gel. For hippocampal extracts, ~200 μg of proteins were
analyzed. Proteins were semi-dry transferred to nitrocellulose and im-
munoblotted using α-Ser3-phosphocolin antibodies (clone 77G2, Cell
Signaling Technology). Blots were stripped and reprobed using α-colin
antibodies (#3312, Cell Signaling Technology or #ACFL02, Cytoskeleton
Inc.).
2.9. Behavioral studies
2.9.1. Animals
All animals were housed at the Center for Experimental Molecular
Medicine at the University of Würzburg, and were kept on a regular 14/
10 h light-dark cycle, in a temperature (21 ± 0.5 °C)- and humidity
(50 ± 5%)-controlled environment with ad libitum access to food and
water. All experiments were performed by the same experienced female
investigator between 9 a.m. and 3 p.m. All mice were males housed in
groups of 25 animals per cage. The age of the mice is specied in the
respective gures. All animal protocols were in line with the provisions
of the Animal Protection Law according to the Directive of the
European Communities Council of 1986 (86/609/EEC). Unless other-
wise indicated, behavioral tests were performed according to standard
procedures using devices from TSE Systems. Mice were imaged with an
infrared light-sensitive CCD camera, and data were collected and ana-
lyzed using the automated tracking software VideoMot2 (TSE Systems).
2.9.2. Tests for anxiety-like behavior, exploratory behavior, and spatial
learning and memory
Mouse behavior in the open eld arena, the elevated plus maze, the
dark-light box and in the Barnes maze was evaluated as described [25].
An independent cohort of mice was analyzed in the Barnes maze; bright
light (~100 lx) was used as a mildly aversive stimulus. Marble burying
was performed as published [26]. Briey, a standard mouse cage
(42.5 × 26.5 × 15 cm) was lled with ~5 cm bedding material. Eigh-
teen glass marbles were spaced evenly in a 3 × 6 grid-fashion on the
surface of the bedding. Mice were placed in the cage for 30 min, and the
number of marbles buried up to 2/3 of their depth was counted.
2.9.3. Sucrose preference test
In a two-bottle free choice test measuring the preferred intake of
either sucrose solution or regular water, mice typically prefer swee-
tened water. A reduction in the sucrose preference ratio in mice is
considered to be indicative of anhedonia, a core symptom of depression
in humans [27]. Mice were presented with two drinking bottles in their
home cage, one containing plain tap water and the other lled with a
2% (w/v) sucrose solution as described elsewhere [27]. The beginning
of the test started with the onset of the dark (active) phase of the ani-
mals' cycle. No food or water deprivation was applied prior to the test.
The position of the two bottles was switched daily to reduce any pos-
sible confounding eect due to a side-bias, and mice were tested for a
total of nine days. The consumption of water and sucrose solution was
estimated simultaneously in control and experimental groups by
weighing the bottles. The sucrose preference ratio was calculated as the
percentage of sucrose intake relative to the total volume of uid intake
using the following equation: Volume (sucrose solution) / [volume
(sucrose solution) + volume (water)] × 100 [28].
2.9.4. Inverted screen test
Each mouse was placed on a standard cage lid and allowed to obtain
arm grip. The lid was then swiftly inverted over an empty mouse cage
lled with bedding. The time until the mouse fell into the empty cage
was measured using a stopwatch; maximum test duration was 5 min.
Mice were given three trials each day with a 20 s recovery phase in
between trials. The test was repeated on three consecutive days. The
cage lid was cleaned before testing another mouse. Latency to fall into
the cage was used to evaluate limb strength.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
3
2.9.5. Weights test
The weights test was performed as described [29], with the excep-
tion that mice were allowed to hold the wire mesh with all four limbs.
Briey, mice were held by the middle of their tails and allowed to grasp
ane wire mesh with an increasing number of steel chain links attached
to it. The mice grasping the wire mesh with the attached weights were
lifted, and the time they held on to the weight was measured with a
stopwatch. The criterion was a 3 s hold; if the mouse dropped the
weight earlier, the test was repeated twice after a recovery phase of
10 s. Failure to hold a given weight three times terminated the trial. If
the mouse succeeded in holding the weight, the next heaviest weight
was tested. The test score indicates the number of links in the heaviest
chain that the mouse held for 3 s multiplied by the time (in s) it held on
to it. For example, a score of 15 indicates a mouse holding a 5-link
weight for 3 s; a score of 16 indicates that it held a 5-link weight for 3 s
and a 6-link weight for 1 s. The weight of each chain was 13 g, the
weight of the mesh attached to the chains links was 5 g.
2.9.6. Grip strength
Hind paw strength was determined using an automated Grip
Strength Meter (Columbus Instruments) as reported previously [30]. In
short, mice were trained to grasp a bar with either their front or hind
paws. A pull was exerted, and the maximal strength that the mice ex-
hibited before releasing the grip bar was measured. For each mouse, ten
measurements per day were acquired on three consecutive days. The
average grip strength value (in N) was determined for each mouse, and
values were expressed relative to the mean values of the respective
control cohort.
2.10. Image quantication and statistical analysis
Western blots were quantied with NIH ImageJ, version 1.45i. For
statistical analysis, two-tailed, unpaired Student's t-tests were per-
formed using GraphPad Prism version 6.0. All samples that were sub-
jected to statistical analysis were biological replicates. The number of
independent samples or individual mice per group and genotype is
specied in each gure or gure legend. Reprobes of Western blot ex-
periments were performed using the same membrane that was probed
for the primary protein under study.
3. Results
3.1. PDXP is a dominant regulator of pyridoxal 5-phosphate levels in vivo
To study the functions of PDXP in vivo, we generated conditionally
Pdxp-decient mice (Supplementary Fig. S1A). Southern blot analysis
demonstrated homologous recombination after breeding Pdxp
x/x
mice
with the whole-body Cre deleter strain EIIa-Cre (Supplementary Fig.
S1B). Homozygous Pdxp-decient (KO) mice on a C57BL/6J back-
ground were born at the expected Mendelian frequencies, and were
indistinguishable from wildtype (WT) controls in terms of viability,
growth and fertility. Fig. 2A demonstrates ecient PDXP deletion in all
investigated organs and cells at the protein level. In accordance with
the important role of the PDXP substrate pyridoxal 5-phosphate (PLP)
for neurotransmitter biosynthesis and metabolism, PDXP was highly
expressed in the brain. The identity of the ~25 kDa band that speci-
cally reacts with the α-PDXP antibody in WT, but not in PDXP-KO
A
37 -
25 -
50 -
37 -
α-a
ctin
α-
PDXP
brain sk. mus. sm. intest. liver
WT KO WT KO WT KO WT KO
brain kidneyheart liver
Ponceau
kDa
37
25
37
WT KO WT KO WT KO WT KO
WT KO WT KO WT KO WT KO
20
15
20
15
kDa
37
25
brain
α-
PDXP
B
C
α-
PDXP
α-
P-cofilin
α-
cofilin
hippocampus
WT KO WT KO WT KO WT KO
α-
PDXP
α-
P-cofilin
α-
cofilin
WT KO
0.0
0.5
1.0
1.5
2.0
relative intensity
P-cofilin/cofilin
p = 0.71
p = 0.34
WT
0
1
2
3
4
20
15
20
15
37
25
relative intensity
P-cofilin/cofilin
kDakDa
>
>
KO
Fig. 2. Characterization of con-
ditionally PDXP-decient mice. (A)
Western blot analysis of PDXP expres-
sion in dierent wildtype (WT) mouse
organs, and loss of detectable PDXP
expression in Pdxp
x/x
; EIIa-Cre mice
(KO). Data are representative of n3
assays performed with tissues dissected
from at least three dierent WT and
three dierent KO mice. The position
of the ~32 kDa PDXP protein is in-
dicated by an arrowhead. The identity
of the ~25 kDa band is currently un-
known. (B, C) Eect of PDXP deletion
on Ser3-phosphocolin (P-colin) le-
vels in whole brain (B) or hippocampal
lysates (C); n= 4 mice per genotype
were analyzed. P-colin signals were
normalized to the respective colin
signals; the densitometric quantica-
tion is shown on the right. The whis-
kers indicate minimum and maximum
values. Samples in (A) were obtained
from 7 to 8 months-old male mice;
samples in (B) and (C) were from
4 months-old male mice. Whole-brain
lysates in (B) and hippocampal lysates
(C) were generated from two separate
mouse cohorts. Sk. mus., skeletal
muscle; sm. intest., small intestine.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
4
brains is currently unclear. Because PDXP isoforms of a corresponding
molecular mass are not known to exist, this band might represent a
proteolytic PDXP degradation product.
After the molecular cloning of PDXP [8], an enzyme termed
chronophin was isolated from bovine brain fractions and shown to
dephosphorylate the actin regulatory protein colin on serine-3 [31];
surprisingly, chronophin and PDXP turned out to be identical. PDXP/
chronophin can regulate colin-dependent actin re-organization at the
leading edge of immune and cancer cells [3235], and mediate an ATP-
sensing mechanism for colin dephosphorylation in neuronal cells ex-
posed to energy-stress [36]. In addition, it has recently been reported
that the constitutive genetic ablation of Pdxp (referred to in that work
as PLPP/CIN) leads to a moderate, ~45% increase in colin phos-
phorylation in the mouse hippocampus; conversely, substantial trans-
genic overexpression of the phosphatase resulted in a ~30% decrease in
colin phosphorylation levels [37]. We analyzed the phosphocolin/
colin ratio in lysates of whole brain (Fig. 2B) and isolated hippocampi
(Fig. 2C) in our mouse model. Phosphocolin levels were more variable
in PDXP-KO than in WT samples, but we did not detect signicant
PDXP-dependent changes.
UPLC-MS/MS-based quantication of PLP, the primary known in
vitro metabolite substrate of PDXP [7,8,38], revealed threefold in-
creases in PLP levels in whole brain extracts of adult PDXP-KO com-
pared to WT mice. PLP levels were also signicantly elevated in skeletal
muscle and in red blood cells. We did not nd PLP changes in liver, in
the small intestine or in plasma (Fig. 3A, Table 1). These ndings in-
dicate that PDXP is not involved in the regulation of extracellular PLP
levels, and suggest that PDXP is not essential for PLP hydrolysis in liver
and intestine. However, in contrast to the brain, liver can release PLP
into the plasma, and secreted PLP can subsequently be hydrolyzed to PL
by alkaline phosphatase, a membrane-bound ectoenzyme. It is therefore
possible that PDXP loss in the liver impacts intracellular PLP levels, but
that this eect is undetectable due to PLP secretion and extracellular
PLP hydrolysis [39]. Likewise, the loss of PDXP in the small intestine
[6] might be compensated for by direct PLP transfer from the intestinal
lumen to the circulation, or by alkaline phosphatase activity [40]. The
lack of increased PLP levels in the plasma of PDXP-KO mice is con-
sistent with a critical role of the extracellular, tissue non-specic al-
kaline phosphatase TNAP for PLP hydrolysis in plasma, as revealed by
the markedly elevated plasma PLP levels measured in patients with
hypophosphatasia, an inborn error characterized by impaired TNAP
activity [41], and by the increased plasma PLP concentrations found in
TNAP-targeted mice (ref. [42] and see below).
Further analysis of the adult brain demonstrated that PDXP loss led
to a comparable PLP increase in all investigated areas (cortex, mid-
brain, hindbrain and cerebellum). We also observed a ~twofold in-
crease in PLP levels in the brains of 6 day-old (postnatal day 6, P6)
PDXP-KO pups (Fig. 3B, Table 2). In brain, PL levels also increased in a
PDXP-dependent manner (Tables 1, 2). However, because the relative
increases in PL and PLP levels were similar in PDXP-KO brain extracts,
we speculate that extracellular TNAP partially hydrolyzed PLP to PL
during cell lysis [42,43]. PN levels in whole-brain extracts of adult
PDXP-KO mice appeared to be increased (Table 1), but the analysis of
dierent adult brain regions or of P6 brains did not conrm this result
(Table 2). By comparison, mice lacking TNAP have reduced PLP levels
in the brain, but strongly elevated PLP levels in plasma (brain: WT,
4.5 ± 1.0 nmol/g wet tissue weight; TNAP-KO, 1.6 ± 0.3 nmol/g wet
tissue weight; serum: WT, 183 ± 104 nM; TNAP-KO, 3166 ±
1288 nM, ref. [42]). Taken together, these data clearly demonstrate
that PDXP is a key determinant in the control of cellular PLP con-
centrations in vivo.
A presumed primary function of PDXP is to keep the levels of free
intracellular PLP low. This is important for cellular homeostasis because
PLP is an electrophilic small molecule that can bind and inactivate
cellular nucleophiles. In addition, elevated PLP levels may impinge
upon cellular PLP biosynthesis. In vitro, PLP functions as an eective
product inhibitor of PNPO, and it additionally binds PNPO tightly at a
non-catalytic site [35,36]. We therefore investigated whether elevated
levels of PLP inuenced PNPO. Western blot analysis of PNPO expres-
sion revealed ~2.4-fold higher PNPO levels in PDXP-KO compared to
WT brain lysates. In contrast, PDXK expression was not signicantly
altered (Fig. 3C). It is possible that higher intracellular PLP levels en-
hance PNPO stability and half-life. Alternatively, elevated PLP levels
might inhibit PNPO activity, and trigger a compensatory increase in
PNPO protein expression.
3.2. Eect of PDXP loss on amino acid levels in the brain
Because PLP is an essential cofactor of many enzymes involved in
amino acid metabolism [1], we next asked whether elevated PLP levels
aected steady-state amino acid levels in the adult brain. As shown in
the Supplementary Table S1, changes were restricted to a subset of
amino acids in particular brain regions. Serine and alanine levels were
signicantly increased in the cortex (+14% or +21%, respectively),
and citrulline levels were higher in the basal ganglia region (+14%).
Glutamine and glycine concentrations decreased in PDXP-KO hind-
brains (20% or 13%, respectively). The levels of pipecolic acid, a
catabolite of lysine, were higher in all parts of the brain, with increases
ranging from +23% in the midbrain to +33% in the cortex. In de-
veloping PDXP-decient brains at P6, alanine and threonine levels were
elevated (+13% or +35%, respectively), and methionine and pheny-
lalanine levels were reduced (35% or 43%, respectively). Together,
these data indicate that higher intracellular PLP concentrations aect
amino acid metabolism, but do not lead to a general increase in the
activities of PLP-dependent enzymes.
3.3. Elevated GABA levels and improved spatial learning and memory in
PDXP-KO mice
PLP is an obligatory cofactor for decarboxylation reactions in the
biosynthesis of dopamine, (nor)epinephrine, serotonin and GABA.
Specically, PLP is required for the aromatic L-amino acid decarbox-
ylase, which catalyzes the decarboxylation of L-3,4-dihydrox-
yphenylalanine (DOPA) to the (nor)epinephrine precursor dopamine,
and the decarboxylation of 5-hydroxy-L-tryptophan to 5-hydro-
xytryptamine (serotonin) [44]. Quantication of these neuro-
transmitters and their catabolites in brain extracts of 9 months-old male
mice did not show PDXP-dependent changes (Fig. 4A). Thus, elevated
cellular PLP concentrations did not increase the physiological activity
of DOPA decarboxylase. Interestingly though, we found signicantly
higher amounts of GABA in whole brain extracts of 8 months-old PDXP-
KO mice, as well as in the cerebellar and midbrain region of 4 months-
old PDXP-KO mice (Fig. 4B). GABA concentrations were also sig-
nicantly higher in whole brains and in dissected cerebellae of P6
PDXP-KO compared to WT pups (Fig. 4B). The rate-limiting step in
GABA biosynthesis consists of the PLP-dependent decarboxylation of
glutamate [45,46]; glutamate itself is formed by glutamate transami-
nase, which also uses PLP as a cofactor. Yet, similar to dopamine,
epinephrine and serotonin, the amounts of glutamate were comparable
between 4 months-old PDXP-KO and WT mice (Fig. 4C). We conclude
that loss of PDXP expression results in a selective increase in cerebral
GABA levels.
One distinguishing feature of GABA biosynthesis is that it is medi-
ated by two glutamate decarboxylases, which are encoded by separate
genes [47]. The majority of GABA in the brain is synthesized by the
67 kDa glutamate decarboxylase (GAD67), a constitutively active, cy-
tosolic enzyme that is typically saturated with PLP. In contrast, the
65 kDa glutamate decarboxylase (GAD65) is enriched in synapses and
bound to the membranes of synaptic vesicles. GAD65 predominantly
exists as an inactive apo-enzyme in cells, and PLP-binding transiently
increases holo-GAD65 formation in response to the demand for extra
GABA in neurotransmission [46,48,49]. Although overall GABA
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
5
p < 0.0001
p = 0.01
4 mo
A
C
0
100
200
300
400
500
0
100
200
300
400
500
cere-
bell.
hind-
brain
cortex mid-
brain
P6
whole
brain
WT
KO
PLP (nmol/g protein)
n.s.
n.s.
p < 0.0001
p < 0.0001
p < 0.0001
p < 0.0001
p = 0.0001
whole
brain
skel.
muscle
liver
sm.
intest.
PLP (nmol/g protein)
WT KO WT KO WT KO
WT KO WT KO WT KO
PDXK PNPO
B
α-PDXK
α-PDXP
α-actin
α-PNPO
α-PDXP
α-actin
0
1
2
3
WT KO WT KO
rel. signal intensity
p = 0.04
n.s.
37 -
37 -
25 -
50 -
37 -
37 -
50 -
25 -
25 -
37 -
kDa
kDa
7-8 mo
Fig. 3. Eect of whole-body PDXP deletion on PLP levels in vivo. PLP levels were quantied by UPLC-MS/MS. (A) Organs were isolated from 7 to 8 months-old male
mice, n=48 per genotype; the indicated brain regions were dissected from 4 months-old male mice, n= 4 per genotype (B). The whiskers indicate minimum and
maximum values. For further details and data on other B6 vitamers, see Tables 1 and 2. (C) Western blot analysis of PDXK and PNPO levels in whole brain lysates of
78 months-old male mice; lysates of n= 3 mice per genotype were analyzed. Right panel, blots were analyzed densitometrically after normalization to actin. WT,
wildtype mice; KO, Pdxp
x/x
; EIIa-Cre mice. Data in (C) are mean values ± S.E.M.; n.s., non-signicant. Skel. muscle, skeletal muscle; sm. intestine, small intestine;
cerebell., cerebellum; P6, postnatal day 6.
Table 1
UPLC-MS/MS-based quantication of B
6
vitamers in WT and PDXP-KO mice.
nmol/g protein nPLP PL PA PMP PM PN
Brain WT 8 91.29 ± 6.30 25.54 ± 1.56 0.27 ± 0.09 156.1 ± 13.6 0.79 ± 0.20 0.06 ± 0.01
PDXP-KO 7 298.8 ± 7.45 74.95 ± 5.95 0.35 ± 0.12 173.6 ± 10.2 1.3 ± 0.24 0.33 ± 0.05
p-Value < 0.0001 < 0.0001 0.60 0.33 0.12 < 0.0001
Skeletal muscle WT 8 235.0 ± 12.0 5.10 ± 1.62 0.05 ± 0.02 75.65 ± 13.17 0.66 ± 0.26 0.01 ± 0.01
PDXP-KO 7 331.4 ± 31.9 8.83 ± 3.40 0.13 ± 0.05 98.69 ± 28.22 0.79 ± 0.35 0.02 ± 0.02
p-Value 0.01 0.32 0.12 0.45 0.77 0.74
Small intestine WT 4 42.75 ± 3.77 7.80 ± 0.66 0.42 ± 0.23 97.25 ± 15.73 0.46 ± 0.12 0.01 ± 0.01
PDXP-KO 4 50.77 ± 3.56 6.62 ± 0.05 0.69 ± 0.12 116.3 ± 3.6 0.71 ± 0.08 0.41 ± 0.17
p-Value 0.17 0.12 0.34 0.28 0.14 0.06
Liver WT 8 152.5 ± 10.9 4.32 ± 0.84 0.68 ± 0.13 222.9 ± 14.3 0.28 ± 0.09 0.024 ± 0.01
PDXP-KO 7 140.9 ± 6.2 3.86 ± 0.56 0.60 ± 0.13 223.7 ± 9.6 0.24 ± 0.06 0.017 ± 0.01
p-Value 0.39 0.66 0.64 0.96 0.70 0.60
nmol/L nPLP PL PA PMP PM PN
Plasma WT 8 146.9 ± 14.9 528.0 ± 45.2 85.88 ± 12.92 246.9 ± 41.2 n.d. n.d.
PDXP-KO 7 143.4 ± 9.3 483.7 ± 33.3 65.14 ± 6.86 142.4 ± 23.2 n.d. n.d.
p-Value 0.85 0.45 0.2 0.05
Red blood cells WT 8 3776 ± 468 393.4 ± 31.4 14.91 ± 1.81 347.4 ± 23.5 n.d. n.d.
PDXP-KO 7 10,321 ± 1402 332.9 ± 23.0 10.20 ± 2.55 389.5 ± 53.2 n.d. n.d.
p-Value < 0.001 0.15 0.15 0.46 ––
Shown are mean values ± S.E.M. Nindicates the number of analyzed mice. For comparison with the other B
6
vitamers, results of the PLP measurements depicted in
Fig. 3A and B are shown again here. Pyridoxine 5-phosphate (PNP) could not be detected. Statistically signicant dierences between the two genotypes were
analyzed using unpaired t-tests. All mice were 78 months-old males; n.d., not determined.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
6
concentrations were increased, we found that GAD67 expression was
lowered by ~30% in PDXP-KO compared to WT brains, possibly as a
result of a negative feedback response (Fig. 4D). GAD65 monomer or
dimer levels were not detectably aected by the absence of PDXP ex-
pression. Interestingly, we observed a prominent high molecular weight
(~220 kDa), GAD65-positive band in immunoblots of PDXP-KO brain
lysates, which was only faintly visible in the controls (Fig. 4D). PLP-
binding to GAD65 has profound eects on protein conformation
[50,51], and elevated PLP levels might thereby favor the formation of
SDS-resistant GAD65 complexes. Taken together, these data suggest
that increased cellular PLP concentrations trigger holo-GAD65 forma-
tion, which augments synaptic GABA biosynthesis in PDXP-KO mouse
brains.
GABA is involved in spatial learning behavior, for example by
regulating adult hippocampal neurogenesis [52,53]. We therefore in-
vestigated the cognitive abilities of PDXP-KO mice in the Barnes maze.
This task reports on hippocampus-dependent spatial reference
memory by evaluating the ability to learn and remember the location
of a hidden escape zone using a set of visual cues [54]. Fig. 5 de-
monstrates that PDXP-KO mice tended to require less time to locate
the escape chamber (reduced target latency), and to search fewer
holes before locating the escape chamber (less primary errors), but
these trends did not reach statistical signicance. However, PDXP-KO
mice required signicantly reduced path lengths (distances) to nd
theescapechambercomparedtoWTmice.PDXP-KOdeletionalsoled
to shorter escape latency, i.e. mice needed less time to enter the escape
chamber. The analysis of reversal learning showed that PDXP-KO mice
performed signicantly better in terms of path length required to nd
the relocated escape chamber and escape latency (Fig. 5). Together,
these results indicate that PDXP loss improved spatial learning and
memory.
3.4. Anxiety-like behavior and impaired motor performance in PDXP-KO
mice
Because of the important roles of GABA for neuronal activities in-
volved in emotion, learning and memory, and the pivotal role of GABA
for muscle tone and the regulation and execution of movements [55],
we further examined PDXP-dependent mouse behavior and motor
performance in a series of non-invasive tests. In the open eld arena (a
sensorimotor test used to assess general activity levels, locomotor ac-
tivity and exploration), PDXP-KO mice spent less time in the center of
the box. This dierence was observed in 45 months old (young) and in
910 months old (middle-aged) mice (Fig. 6A). In the elevated plus
maze (a test used to evaluate anxiety-related behavior), young PDXP-
decient mice behaved similar to WT controls, whereas middle-aged
PDXP-KO mice entered the open arms less frequently and also stayed
there for shorter times (Fig. 6B). Analysis of dark/light exploration
showed a tendency towards anxiety-like behavior in PDXP-KO mice (as
indicated by an increased latency to enter the lit compartment). How-
ever, this trend did not reach statistical signicance; likewise, the time
spent in the lit compartment was not dierent between WT and KO
mice (Fig. 6C). Marble burying (a test that reports on anxiety-like,
obsessive-compulsive, and repetitive behavior) increased in older
PDXP-KO compared to age-matched WT mice (Fig. 6D). General loco-
motor activity (measured for all mice in parallel with the parameters
shown in Fig. 6AC) was not dierent between the two genotypes in
these tasks. A sucrose preference test was conducted in 9 months-old
male mice to evaluate anhedonia as an indicator of depressive-like
behavior, yet no PDXP-dependent dierences were observed (% sucrose
intake relative to total uid intake in mL: WT, 93.70 ± 2.08 mL, n=3;
KO, 88.70 ± 5.15 mL, n=3;p= 0.42. Data are means ± S.E.M.). We
conclude from these results that PDXP loss resulted in a mild, anxiety-
like phenotype that was more prominent in middle-aged than in
younger mice.
We next analyzed PDXP-dependent motor function. To exclude
potential body weight dierences as a confounding factor, we rst
compared the body weights of 45 months-old and 89 months-old
male mice. There was a trend towards higher body weights in middle-
aged, whole-body PDXP-KO mice, but this tendency did not reach sta-
tistical signicance (45 months-old: WT, 29.64 ± 0.30 g, n=8;
PDXP-KO; 30.18 ± 1.06 g, n=8, p= 0.63; 89 months-old: WT,
31.23 ± 0.63 g, n= 12; PDXP-KO, 33.36 ± 0.86 g, n= 12, p= 0.06.
Data are means ± S.E.M.). The inverted screen test showed a sig-
nicantly decreased performance of older PDXP-KO compared to WT
mice (Fig. 7A). In the weights test, performance was already impaired
in younger PDXP-KO mice (Fig. 7B; see Experimental Procedures for
details). To evaluate muscle strength directly, we conducted grip
strength measurements. Fore paw grip strength measurements in
89 months-old male mice did not show PDXP-dependent dierences
(WT 1.46 ± 0.04 N, n= 12; KO 1.42 ± 0.07 N, n= 11, p= 0.61). In
contrast, hind paw grip strength measurements revealed poorer per-
formance of PDXP-KO mice (WT 0.92 ± 0.02 N, n=8; KO
0.77 ± 0.02 N, n= 11; p< 0.0001. Data for front and hind paw
measurements are means ± S.E.M.).
PLP is a cofactor of glycogen phosphorylase, the enzyme that cat-
alyzes the rate-limiting step in glycogenolysis. Because muscle glycogen
is an important energy substrate during exercise, and PLP levels in
skeletal muscle were signicantly increased in PDXP-KO mice (see
Fig. 3A), we asked whether PDXP loss in skeletal muscle or in the
Table 2
UPLC-MS/MS-based quantication of B
6
vitamers in the brains of adult and postnatal WT and PDXP-KO mice.
nmol/g protein nPLP PL PA PMP PM PN
Cerebellum WT 4 85.10 ± 7.05 9.18 ± 0.69 0.032 ± 0.03 162.1 ± 22.46 0.14 ± 0.02 0.01 ± 0.01
PDXP-KO 4 266.9 ± 11.31 19.91 ± 3.82 0.12 ± 0.03 193.2 ± 18.26 0.14 ± 0.03 0.04 ± 0.02
p-Value < 0.0001 0.03 0.1 0.32 0.95 0.22
Hindbrain WT 4 89.08 ± 8.08 18.17 ± 1.99 n.d. 225.0 ± 7.200 0.55 ± 0.22 0.04 ± 0.02
PDXP-KO 4 234.8 ± 14.02 44.23 ± 1.97 n.d. 234.5 ± 14.82 0.63 ± 0.08 0.07 ± 0.05
p-Value 0.0001 < 0.0001 0.59 0.75 0.58
Cortex WT 4 64.33 ± 3.89 13.16 ± 1.15 0.09 ± 0.03 145.1 ± 13.59 0.37 ± 0.09 0.007 ± 0.01
PDXP-KO 4 250.8 ± 18.15 34.53 ± 3.07 0.16 ± 0.07 158.7 ± 10.24 0.38 ± 0.10 0.06 ± 0.03
p-Value < 0.0001 < 0.001 0.41 0.46 0.97 0.10
Midbrain WT 4 85.41 ± 0.17 15.35 ± 1.25 0.06 ± 0.04 155.8 ± 15.03 0.39 ± 0.11 0.010 ± 0.004
PDXP-KO 4 277.8 ± 6.02 43.18 ± 2.94 0.04 ± 0.04 191.1 ± 6.6 0.74 ± 0.17 0.11 ± 0.05
p-Value < 0.0001 0.0001 0.73 0.07 0.14 0.09
P6 brain WT 4 95.25 ± 6.82 28.91 ± 1.21 0.07 ± 0.05 51.39 ± 4.98 0.17 ± 0.05 n.d.
PDXP-KO 4 201.8 ± 3.97 48.00 ± 1.63 0.04 ± 0.04 53.53 ± 8.86 0.26 ± 0.03 0.01 ± 0.01
p-Value < 0.0001 < 0.0001 0.65 0.84 0.18 0.36
Shown are mean values ± S.E.M. Nindicates the number of analyzed mice. For comparison with the other B
6
vitamers, results of the PLP measurements depicted in
Fig. 3A and B are shown again here. Pyridoxine 5-phosphate (PNP) could not be detected. Statistically signicant dierences between the two genotypes were
analyzed using unpaired t-tests. Adult mice were 4 months-old males. P6, postnatal day 6; n.d., not detected.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
7
A
whole brain
p < 0.001
p < 0.01 p < 0.01
p = 0.02
p < 0.001
B
0
10
20
30
40
50
60
70
0
50
100
150
200
250
adult P6
whole brain
cerebellum
cerebellum
hindbrain
cortex
midbrain
glutamate (µmol/g protein)
Cadult P6
whole brain
cerebellum
cerebellum
hindbrain
cortex
midbrain
n.s.
n.s.
GABA (µmol/g protein)
n.s.
n.s.
n.s.
n.s.
n.s. n.s.
DA
DOPAC
epinephr.
HVA
5-HT
5-HIAA
0.0
0.5
1.0
1.5
3
6
9n.s.
n.s.
n.s. n.s.
n.s.
n.s.
neurotransmitter/metabolite
(nmol/g wet brain)
WT
KO
α-
GAD 67
WT KO WT KO WT KO
150 -
100 -
kDa
250 -
150 -
100 -
75 -
50 -
WT KO WT KO WT KO WT KOWT KO
-
-
-
-
-
-
-
α-
GAD 65
α-
FAK
α-
FAK
p = 0.001 n.s.
p < 0.001
rel. signal intensity
WT KO WT KO
WT KO
<<
<<
GAD67 GAD65
GAD65 HMW
rel. signal intensity
0
5
10
15
0.00
0.25
0.50
0.75
1.00
1.25
D
Fig. 4. Eect of whole-body PDXP deletion on neurotransmitter levels in the brain. (A) HPLC-based quantication of the indicated neurotransmitters and their
catabolites in whole-brain extracts of 9 months-old male mice (n= 5 per genotype). DA, dopamine; DOPAC, 3,4-dihydroxyphenyl acetic acid; HVA, homovanillic
acid; epinephr., epinephrine; 5-HT, 5-hydroxytryptamine (serotonin); 5-HIAA, 5-hydroxyindole acetic acid. (B) UPLC-MS/MS-based quantication of GABA levels in
the brain. Whole brains were isolated from 8 months-old male mice (n= 4 per genotype). The indicated brain regions were dissected from 4 months-old male mice
(n= 4 per genotype). P6, analysis of whole brains or dissected cerebellae from pups at postnatal day 6 (n= 4 per genotype). (C) UPLC-MS/MS-based quantication
of glutamate levels in the brain. Samples were the same as those analyzed in (B). The whiskers indicate minimum and maximum values. (D) PDXP deciency aects
GAD67 and GAD65. Left and middle panels, GAD67 and GAD65 immunoblots. Brain lysates were generated from 8 months-old male mice, n= 4 per genotype. Focal
adhesion kinase (FAK) served as a loading control. Blots were analyzed densitometrically, and GAD65/67 signals were normalized to FAK signals. The top right panel
shows the quantication of GAD monomers (indicated by <in the blots), and the bottom right panel of the GAD65 high-molecular weight (HMW) complexes
(indicated by <<in the blots). Note that all bands corresponding to GAD monomers and HMW complexes were analyzed densitometrically and are included in
the gure, but that some symbols are overlapping and thus hidden. Data are mean values ± S.E.M.; n.s., non-signicant. WT, wildtype mice; KO, Pdxp
x/x
; EIIa-Cre
mice.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
8
nervous system was primarily responsible for the apparent motor im-
pairments. To this end, mice decient for PDXP either in skeletal
muscle (Pdxp
x/x
; Acta1-Cre
+/
; mKO) or in neural cells (Pdxp
x/x
;
Nestin-Cre
+/
; nKO) were generated. We veried ecient PDXP dele-
tion in skeletal muscle and whole brain extracts in these two lines
(Supplementary Fig. S2A, B). Because of the known metabolic issues
related to knockouts driven by Nestin-Cre [56], mice with a neural cell-
directed PDXP deletion were only characterized at 45 months of age.
PDXP deletion in neural cells led to an increase in whole brain PLP
levels (Supplementary Fig. S2C). Although there was a tendency to-
wards increased GABA concentrations in nKO mice, these changes were
statistically non-signicant (Supplementary Fig. S2D). Similar to brain
extracts generated from whole-body PDXP-KO mice (see Fig. 4D), we
detected a high molecular weight, GAD65-positive band in im-
munoblots of nKO brain lysates (Supplementary Fig. S2E). These results
suggest that the appearance of high molecular weight GAD65 com-
plexes is a sensitive indicator of increased PLP levels, and that cell types
other than neurons, for example glial and endothelial cells [57] con-
tribute to the overall regulation of GABA levels in whole-body PDXP-KO
mice.
Grip strength measurements in young mice showed that PDXP ab-
lation in neural cells signicantly reduced hind paw strength, whereas
PDXP loss in skeletal muscle increased hind paw strength in older mice
(Fig. 7C). Hence, PDXP deciency in skeletal muscle does not account
for the decreased grip strength of globally PDXP-decient mice. We
conclude that a dominant loss of PDXP function in another organ, most
likely the brain, causes the decrease in neuromuscular strength ob-
served in whole-body PDXP-KO mice.
4. Discussion
We demonstrate that PDXP (also known as PLPP, chronophin or
CIN) is a major regulator of PLP concentrations in vivo, and that whole-
body PDXP ablation elevates PLP and GABA levels in the brain.
Spatial learning and memory was improved in PDXP-decient
compared to WT mice, and GABA may be involved in this behavior.
Many neuropsychiatric disorders and neurodegenerative diseases en-
compass dysfunctional GABA system components, and the disruption of
GABAergic inhibitory circuits with a resulting excitatory/inhibitory
imbalance is likely to contribute to some of the clinical features of these
disorders [5860]. GABA signaling is also impaired in the aged hip-
pocampus, suggesting that pharmacological strategies might improve
memory function and spatial navigation abilities in the elderly popu-
lation [61,62]. A recent neuroimaging study that investigated the as-
sociation between vitamin B6 blood levels and cognition, brain struc-
ture and functional connectivity in healthy older humans (N > 600;
age range 5585 years) has shown that vitamin B6 supplementation
was positively related to cortical folding and thus to the preservation of
brain structure. However, neuropsychological testing did not detect an
improvement in cognitive performance, and only slight changes in
functional connectivity were found [63]. Based on these data, the au-
thors hypothesized that vitamin B6 might counteract the gray matter
loss that frequently occurs in older age, and stabilize cognitive abilities.
A meta-analysis of brain GABA levels in dierent neuropsychiatric
diseases has revealed a GABAergic decit in the brains of subjects with
autism spectrum disorders and major depression [64], and subtype-
selective GABA-A receptor modulators show potential as novel anti-
depressants [65,66]. GABA concentrations below a certain threshold
lead to seizures, and a number of anti-epileptic drugs target the GA-
BAergic system [67]. GABA levels are determined by GABA biosynth-
esis via PLP-dependent glutamic acid decarboxylases, and GABA cata-
bolism through the PLP-dependent GABA transaminase. The selective
GABA transaminase suicide inhibitor vigabatrin [68] increases GABA
levels, and is used for the treatment of infantile spasms and as an ad-
junctive therapy for refractory and complex partial seizures in adults
[69,70]. Despite its clinical ecacy, vigabatrin therapy is associated
with harmful side eects, such as the dose-dependent risk for pro-
gressive and permanent visual eld defects [69,70]. It is tempting to
speculate that a combination of a pharmacological PDXP inhibitor to-
gether with vigabatrin may further increase GABA levels in the brain
and allow for a vigabatrin dose reduction.
The anxiety-like phenotype of PDXP-KO mice was unexpected,
given the physiological role of GABA in counteracting anxiety via
GABA-A receptors [58,65,66]. However, PLP and GABA levels were
already elevated postnatally in our mouse model. GABA levels aect
brain development [71] and synaptogenesis [72], and can thereby
impact stress responses later in life. For example, postnatal adminis-
tration of the GABA-A receptor agonist muscimol results in anxiety-like
behavior in 3-months old mice [73]. The EIIa-Cre driven, embryonic
ablation of PDXP might aect brain development (through GABA and/
or other mechanisms), and inuence stress susceptibility and anxiety in
that way. PDXP loss might also alter the balance between excitatory and
inhibitory transmission in specic synapses in the adult organism. For
example, GABA co-released with glutamate controls the activity of the
lateral habenula, a region in the dorsal thalamus that regulates mood
and anxiety [74]. Finally, we found elevated D/L-serine levels (stereo-
isomers were not resolved in our analysis) in the cortex, and decreased
glycine concentrations in the hindbrains of PDXP-KO mice. Serine ra-
cemase is a PLP-dependent enzyme, and D-serine is a co-agonist at N-
methyl-D-aspartic acid (NMDA) glutamate receptors in the brain. Gly-
cine acts as an inhibitory neurotransmitter in the spinal cord and
brainstem, and has excitatory eects in the cerebral cortex due to its
agonistic activity at glutamatergic NMDA receptors. These changes may
contribute to the observed behavioral phenotypes. Further work is ne-
cessary to investigate the electrophysiological consequences of PDXP
1234567
0
5
10
15
three-trial blocks
primary errors (#)
1234567
0
20
40
60
80
100
three-trial blocks
1234567
0
50
100
150
three-trial blocks
1234567
0
200
400
600
800
1000
three-trial blocks
distance (cm)
KO
WT
target latency (sec)escape latency (sec)
p=0.026
p<0.001
p=0.006
p=0.007
p=0.008
p=0.007
p=0.004
p=0.013
p=0.027
p=0.030
Fig. 5. Whole-body PDXP loss improves performance in the Barnes maze.
Spatial learning and memory was assessed over ve consecutive days, and three
trials were performed per day (three-trial blocks, shown as mean
values ± S.E.M.). Afterwards, reversal learning was tested by moving the es-
cape chamber to the opposite side of the maze (indicated by the vertical dotted
line), and mice were analyzed for another two days in six additional trials.
Scored parameters were target latency (time to locate the hole with the escape
chamber), primary errors (number of holes searched before nding the hole
equipped with the escape chamber), escape latency (time needed to enter the
escape chamber) and distance (overall path length required before reaching the
hole with the escape chamber). Tests were performed with n= 12 mice per
genotype. All mice were 7 months-old males. Data are mean values ± S.E.M.,
p-values indicate statistically signicant dierences between WT and KO mice
in each trial block. WT, wildtype mice; KO, Pdxp
x/x
; EIIa-Cre mice.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
9
loss in specic brain regions and synapses, and to characterize the in-
volved receptors and signaling pathways in our mouse model [75].
In addition to a GABA-dependent decrease in muscle tone [76], the
apparent motor decits of PDXP-KO mice might be inuenced by in-
creased anxiety. In addition, age-dependent processes related to con-
tinuously elevated cellular PLP levels may contribute to the observed
phenotype. An increased anxiety-like behavior could add to the retarda-
tion of spatial learning that was observed when PLPP/CIN (PDXP)-de-
cient mice were tested in the Morris water maze in another study [37].
The water maze imposes a much stronger aversive stimulus and more
stressful conditions on mice than the dry-land Barnes maze used in our
study, and increased stress and anxiety may confound the assay outcome
[54]. The discrepant results obtained in the two PDXP-KO mouse models
might also be linked to dierences in the analyzed mouse strains, as well
as in animal age, -sex or -housing conditions. The mice investigated in our
present study were conditionally PDXP-decient males of the indicated
ages; animals were on a C57BL/6J background and maintained under
specic pathogen-free conditions. Kim et al. have described mice with a
constitutive PDXP knockout on a mixed 129/SvEv-C57BL/6J albino
background; age, sex and housing conditions of the animals were not
A
C
D
0.0
0.5
1.0
1.5
2.0
0
10
20
30
40
50
B
0
10
20
30
40
p=0.002
0
20
40
60
80
p<0.0001
p=0.034
0
10
20
30 p=0.010
p=0.015
overall center time (%)
WT KO KO
WT
4-5 mo 9-10 mo
time on open arms (%)
WT KO KOWT
4-5 mo 9-10 mo
open arm entries (%)
WT KO KOWT
4-5 mo 9-10 mo
time in lit compartment (%)
latency (min)
WT KO KO
WT
4-5 mo 9-10 mo
WT KO KO
WT
4-5 mo 9-10 mo
0
5
10
15
20
marbles buried (#)
WT KO KOWT
4-5 mo 9-10 mo
p=0.016
n.s.
p=0.001
p=0.042
812128
812128
812128812128
8121288 12128
n.s.
n.s. n.s.
n.s.
0
500
1000
1500
2000
2500
distance (cm)
0
1500
3000
4500
6000
distance (cm) distance (cm)
0
500
1000
1500
2000
2500
WT KO KOWT
4-5 mo 9-10 mo
812128
WT KO KO
WT
4-5 mo 9-10 mo
812128
WT KO KOWT
4-5 mo 9-10 mo
812128
Fig. 6. Whole-body PDXP loss results in a
mild anxiety-like behavior. (A) Open eld
test. Scored parameters were time in the
center of the open eld arena and overall
locomotion (horizontal distance covered in
the 30 min test period). (B) Elevated plus
maze. Open arm entries, time on open arms
and overall locomotion were scored during
the 10 min test period. (C) Dark/light box.
The latency to enter and the time spent in
the lit compartment was measured together
with the overall locomotion in the 10 min
test period. (D) Marble burying. The
number of marbles buried within the 30 min
test period was counted. Mo, months. Data
are mean values + S.E.M. The number of
analyzed mice is indicated in the bars. WT,
wildtype mice; KO, Pdxp
x/x
; EIIa-Cre mice.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
10
reported [37]. The inbred strain background can impact behavioral
measures, and learning and memory is an age-dependent process [7779].
Previous work by us and others has investigated the colin reg-
ulatory function of PDXP [3136], and a recent study has reported ef-
fects of PDXP on hippocampal colin phosphoregulation in vivo [37].
Yet, a direct colin phosphatase activity of PDXP is dicult to reconcile
with crystallographic work from dierent laboratories, including our
own [21,38,80] (Protein Data Bank entries 2OYC, 2P69, 2P27, 2CFR,
2CFS, 2CFT, 5GYN, 4BX3, 4BXO, 4BX2, 4BKM, 5AES). PLP is deeply
buried within the catalytic cleft of PDXP [80], and the active site is
occluded by a large capping domain typical of small molecule-directed,
haloacid dehalogenase (HAD)-type hydrolases [81,82]. Given these
structural features, extensive conformational changes would be re-
quired to accommodate a protein substrate. Without structural in-
formation on the mechanism of colin dephosphorylation by PDXP, it
currently appears more likely that the eects of PDXP on phosphoco-
lin that are found in some studies are caused by indirect mechanisms.
Supplementary data to this article can be found online at https://
doi.org/10.1016/j.bbadis.2018.08.018.
Transparency document
The Transparency document associated this article can be found, in
online version.
Acknowledgements
We thank Angelika Keller and Kerstin Hadamek for excellent tech-
nical assistance, and Heinrich Blazyca and Drs. Dennis Klein and Rudolf
Martini for generous help with grip strength measurements. Marjolein
Bosma is thanked for the analysis of B6 vitamers. We acknowledge the
assistance of Charlotte Auth and Gabriel Christmann with immunoblots.
This work was supported by the Deutsche Forschungsgemeinschaft
SFB728 and SFB688 (to AG) and the IZKF Würzburg (to LH). CW was
supported by a predoctoral fellowship from the Medical Faculty of the
University of Würzburg (Graduate School of Life Sciences) and by a
Kaltenbach predoctoral fellowship from the German Heart Foundation.
References
[1] R. Percudani, A. Peracchi, A genomic overview of pyridoxal-phosphate-dependent
enzymes, EMBO Rep. 4 (2003) 850854.
[2] J.P. Richard, T.L. Amyes, J. Crugeiras, A. Rios, Pyridoxal 5-phosphate: electrophilic
catalyst extraordinaire, Curr. Opin. Chem. Biol. 13 (2009) 475483.
[3] M.L. di Salvo, R. Contestabile, M.K. Safo, Vitamin B(6) salvage enzymes: me-
chanism, structure and regulation, Biochim. Biophys. Acta 1814 (2011) 15971608.
[4] N. Darin, E. Reid, L. Prunetti, L. Samuelsson, R.A. Husain, M. Wilson, B. El Yacoubi,
E. Footitt, W.K. Chong, L.C. Wilson, H. Prunty, S. Pope, S. Heales, K. Lascelles,
M. Champion, E. Wassmer, P. Veggiotti, V. de Crecy-Lagard, P.B. Mills,
P.T. Clayton, Mutations in PROSC disrupt cellular pyridoxal phosphate homeostasis
and cause vitamin-B6-dependent epilepsy, Am. J. Hum. Genet. 99 (2016)
13251337.
[5] J.W. Whittaker, Intracellular tracking of the pyridoxal cofactor. Implications for
health and metabolic disease, Arch. Biochem. Biophys. 592 (2016) 2026.
[6] M. Albersen, M. Bosma, N.V. Knoers, B.H. de Ruiter, E.F. Diekman, J. de Ruijter,
W.F. Visser, T.J. de Koning, N.M. Verhoeven-Duif, The intestine plays a substantial
role in human vitamin B6 metabolism: a Caco-2 cell model, PLoS One 8 (2013)
e54113.
[7] M.L. Fonda, Purication and characterization of vitamin B6-phosphate phosphatase
from human erythrocytes, J. Biol. Chem. 267 (1992) 1597815983.
[8] Y.M. Jang, D.W. Kim, T.C. Kang, M.H. Won, N.I. Baek, B.J. Moon, S.Y. Choi,
O.S. Kwon, Human pyridoxal phosphatase. Molecular cloning, functional expres-
sion, and tissue distribution, J. Biol. Chem. 278 (2003) 5004050046.
[9] M.L. di Salvo, M.K. Safo, R. Contestabile, Biomedical aspects of pyridoxal 5-
phosphate availability, Front. Biosci. (Elite Ed.) 4 (2012) 897913.
[10] P. Zhang, K. Tsuchiya, T. Kinoshita, H. Kushiyama, S. Suidasari, M. Hatakeyama,
H. Imura, N. Kato, T. Suda, Vitamin B6 prevents IL-1beta protein production by
inhibiting NLRP3 inammasome activation, J. Biol. Chem. 291 (2016)
2451724527.
[11] P.M. Ueland, A. McCann, O. Midttun, A. Ulvik, Inammation, vitamin B6 and re-
lated pathways, Mol. Asp. Med. 53 (2017) 1027.
[12] P.T. Clayton, B6-responsive disorders: a model of vitamin dependency, J. Inherit.
Metab. Dis. 29 (2006) 317326.
[13] P.B. Mills, E. Struys, C. Jakobs, B. Plecko, P. Baxter, M. Baumgartner,
M.A. Willemsen, H. Omran, U. Tacke, B. Uhlenberg, B. Weschke, P.T. Clayton,
Mutations in antiquitin in individuals with pyridoxine-dependent seizures, Nat.
Med. 12 (2006) 307309.
[14] P.B. Mills, R.A. Surtees, M.P. Champion, C.E. Beesley, N. Dalton, P.J. Scambler,
S.J. Heales, A. Briddon, I. Scheimberg, G.F. Homann, J. Zschocke, P.T. Clayton,
Neonatal epileptic encephalopathy caused by mutations in the PNPO gene encoding
pyridox(am)ine 5-phosphate oxidase, Hum. Mol. Genet. 14 (2005) 10771086.
[15] G.F. Homann, B. Schmitt, M. Windfuhr, N. Wagner, H. Strehl, S. Bagci, A.R. Franz,
P.B. Mills, P.T. Clayton, M.R. Baumgartner, B. Steinmann, T. Bast, N.I. Wolf,
J. Zschocke, Pyridoxal 5-phosphate may be curative in early-onset epileptic en-
cephalopathy, J. Inherit. Metab. Dis. 30 (2007) 9699.
[16] M. Albersen, M. Bosma, J.J. Jans, F.C. Hofstede, P.M. van Hasselt, M.G. de Sain-van
der Velden, G. Visser, N.M. Verhoeven-Duif, Vitamin B6 in plasma and cere-
brospinal uid of children, PLoS One 10 (2015) e0120972.
[17] B. Plecko, M. Zweier, A. Begemann, D. Mathis, B. Schmitt, P. Striano,
M. Baethmann, M.S. Vari, F. Beccaria, F. Zara, L.M. Crowther, P. Joset, H. Sticht,
S.M. Papuc, A. Rauch, Conrmation of mutations in PROSC as a novel cause of
vitamin B6-dependent epilepsy, J. Med. Genet. 54 (2017) 809814.
[18] H. Schaumburg, J. Kaplan, A. Windebank, N. Vick, S. Rasmus, D. Pleasure,
M.J. Brown, Sensory neuropathy from pyridoxine abuse. A new megavitamin syn-
drome, N. Engl. J. Med. 309 (1983) 445448.
[19] K. Dalton, M.J. Dalton, Characteristics of pyridoxine overdose neuropathy syn-
drome, Acta Neurol. Scand. 76 (1987) 811.
[20] S.M. Gospe Jr., M.P. Adam, H.H. Ardinger, R.A. Pagon, S.E. Wallace, L.J.H. Bean,
K. Stephens, A. Amemiya (Eds.), Pyridoxine-dependent Epilepsy, GeneReviews®,
Seattle (WA), 1993.
[21] G. Knobloch, N. Jabari, S. Stadlbauer, H. Schindelin, M. Kohn, A. Gohla, Synthesis
of hydrolysis-resistant pyridoxal 5-phosphate analogs and their biochemical and X-
ray crystallographic characterization with the pyridoxal phosphatase chronophin,
Bioorg. Med. Chem. 23 (2015) 28192827.
[22] H. Prinsen, B.G.M. Schiebergen-Bronkhorst, M.W. Roeleveld, J.J.M. Jans,
M.G.M. de Sain-van der Velden, G. Visser, P.M. van Hasselt, N.M. Verhoeven-Duif,
AB
0
5
10
15
20
25 p=0.001
p=0.012
0
1
2
3
4
WT KO WT
4-5 mo 9-10 mo
p<0.0001
p<0.0001
test score
time (min)
0.6
0.8
1.0
1.2
1.4
hindpaw strength
(normalized to mean of controls)
p=0.004
p=0.039p=0.007
n.s.
KO WT KO WT
4-5 mo 9-10 mo
KO
24 24 36 36 88 12 12
C
4-5 mo 9-10 mo
WT KO WT KO WT mKOnKOWT
89 97 1211 66
Fig. 7. PDXP loss impairs motor performance and reduces muscle strength. (A)
Inverted screen test. Latency to fall othe grid into the cage was measured. (B)
Weights test. The test score represents the number of chain links the mice lifted
for 3 s, multiplied by the time the weight was held in sec. Data in (A) and (B) are
mean values + S.E.M. (C) Hind paw grip strength measurements. Grip strengths
were normalized to the mean values of the respective control cohorts. The
whiskers show minimum and maximum values; the median value is indicated in
the box. The number of analyzed mice are given in the bars or underneath the
box plots. KO, whole-body PDXP-KO; nKO, neural cell-specic PDXP-KO; mKO,
skeletal muscle-specic PDXP-KO. All animals were males; WT signies oxed
littermate controls (for nKO and mKO mice) or WT mice (for whole-body KO).
Mo, months; n.s., non-signicant.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
11
Rapid quantication of underivatized amino acids in plasma by hydrophilic inter-
action liquid chromatography (HILIC) coupled with tandem mass-spectrometry, J.
Inherit. Metab. Dis. 39 (2016) 651660.
[23] R.R. Schur, M.P. Boks, E. Geuze, H.C. Prinsen, N.M. Verhoeven-Duif, M. Joels,
R.S. Kahn, E. Vermetten, C.H. Vinkers, Development of psychopathology in de-
ployed armed forces in relation to plasma GABA levels, Psychoneuroendocrinology
73 (2016) 263270.
[24] M. van der Ham, M. Albersen, T.J. de Koning, G. Visser, A. Middendorp, M. Bosma,
N.M. Verhoeven-Duif, M.G. de Sain-van der Velden, Quantication of vitamin B6
vitamers in human cerebrospinal uid by ultra performance liquid chromato-
graphy-tandem mass spectrometry, Anal. Chim. Acta 712 (2012) 108114.
[25] A. Raab, S. Popp, K.P. Lesch, M.J. Lohse, M. Fischer, J. Deckert, L. Hommers,
Increased fear learning, spatial learning as well as neophobia in Rgs2(/) mice,
Genes Brain Behav. 17 (2018) e12420.
[26] R.M. Deacon, Digging and marble burying in mice: simple methods for in vivo
identication of biological impacts, Nat. Protoc. 1 (2006) 122124.
[27] T. Strekalova, R. Spanagel, D. Bartsch, F.A. Henn, P. Gass, Stress-induced anhedonia
in mice is associated with decits in forced swimming and exploration,
Neuropsychopharmacology 29 (2004) 20072017.
[28] M.Y. Liu, C.Y. Yin, L.J. Zhu, X.H. Zhu, C. Xu, C.X. Luo, H. Chen, D.Y. Zhu,
Q.G. Zhou, Sucrose preference test for measurement of stress-induced anhedonia in
mice, Nat. Protoc. 13 (2018) 16861698.
[29] R.M. Deacon, Measuring the strength of mice, J. Vis. Exp. 76 (2013), https://doi.
org/10.3791/2610.
[30] B. Kohl, S. Fischer, J. Groh, C. Wessig, R. Martini, MCP-1/CCL2 modies axon
properties in a PMP22-overexpressing mouse model for Charcot-Marie-tooth 1A
neuropathy, Am. J. Pathol. 176 (2010) 13901399.
[31] A. Gohla, J. Birkenfeld, G.M. Bokoch, Chronophin, a novel HAD-type serine protein
phosphatase, regulates colin-dependent actin dynamics,Nat.CellBiol.7(2005)2129.
[32] M. Schulze, O. Fedorchenko, T.G. Zink, C.B. Knobbe-Thomsen, S. Kraus, S. Schwinn,
A. Beilhack, G. Reifenberger, C.M. Monoranu, A.L. Siren, E. Jeanclos, A. Gohla,
Chronophin is a glial tumor modier involved in the regulation of glioblastoma
growth and invasiveness, Oncogene 35 (2016) 31633177.
[33] V. Delorme-Walker, J.Y. Seo, A. Gohla, B. Fowler, B. Bohl, C. DerMardirossian,
Chronophin coordinates cell leading edge dynamics by controlling active colin
levels, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) E5150E5159.
[34] C.X. Sun, M.A. Magalhaes, M. Glogauer, Rac1 and Rac2 dierentially regulate actin
free barbed end formation downstream of the fMLP receptor, J. Cell Biol. 179
(2007) 239245.
[35] M. Zoudilova, P. Kumar, L. Ge, P. Wang, G.M. Bokoch, K.A. DeFea, Beta-arrestin-
dependent regulation of the colin pathway downstream of protease-activated re-
ceptor-2, J. Biol. Chem. 282 (2007) 2063420646.
[36] T.Y. Huang, L.S. Minamide, J.R. Bamburg, G.M. Bokoch, Chronophin mediates an
ATP-sensing mechanism for colin dephosphorylation and neuronal colin-actin
rod formation, Dev. Cell 15 (2008) 691703.
[37] J.E. Kim, Y.J. Kim, D.S. Lee, J.Y. Kim, A.R. Ko, H.W. Hyun, M.J. Kim, T.C. Kang,
PLPP/CIN regulates bidirectional synaptic plasticity via GluN2A interaction with
postsynaptic proteins, Sci. Rep. 6 (2016) 26576.
[38] C. Kestler, G. Knobloch, I. Tessmer, E. Jeanclos, H. Schindelin, A. Gohla,
Chronophin dimerization is required for proper positioning of its substrate speci-
city loop, J. Biol. Chem. 289 (2014) 30943103.
[39] S.P. Coburn, Vitamin B-6 metabolism and interactions with TNAP, Subcell.
Biochem. 76 (2015) 207238.
[40] H. Mehansho, M.W. Hamm, L.M. Henderson, Transport and metabolism of pyr-
idoxal and pyridoxal phosphate in the small intestine of the rat, J. Nutr. 109 (1979)
15421551.
[41] M.P. Whyte, J.D. Mahuren, L.A. Vrabel, S.P. Coburn, Markedly increased circulating
pyridoxal-5-phosphate levels in hypophosphatasia. Alkaline phosphatase acts in
vitamin B6 metabolism, J. Clin. Invest. 76 (1985) 752756.
[42] K.G. Waymire, J.D. Mahuren, J.M. Jaje, T.R. Guilarte, S.P. Coburn, G.R. MacGregor,
Mice lacking tissue non-specic alkaline phosphatase die from seizures due to de-
fective metabolism of vitamin B-6, Nat. Genet. 11 (1995) 4551.
[43] H. Zimmermann, D. Langer, Tissue-nonspecic alkaline phosphatase in the devel-
oping brain and in adult neurogenesis, Subcell. Biochem. 76 (2015) 6184.
[44] M.K. Rahman, T. Nagatsu, T. Sakurai, S. Hori, M. Abe, M. Matsuda, Eect of pyr-
idoxal phosphate deciency on aromatic L-amino acid decarboxylase activity with
L-DOPA and L-5-hydroxytryptophan as substrates in rats, Jpn. J. Pharmacol. 32
(1982) 803811.
[45] E. Roberts, K. Kuriyama, Biochemical-physiological correlations in studies of the
gamma-aminobutyric acid system, Brain Res. 8 (1968) 135.
[46] D.L. Martin, K. Rimvall, Regulation of gamma-aminobutyric acid synthesis in the
brain, J. Neurochem. 60 (1993) 395407.
[47] D.F. Bu, M.G. Erlander, B.C. Hitz, N.J. Tillakaratne, D.L. Kaufman, C.B. Wagner-
McPherson, G.A. Evans, A.J. Tobin, Two human glutamate decarboxylases, 65-kDa
GAD and 67-kDa GAD, are each encoded by a single gene, Proc. Natl. Acad. Sci. U.
S. A. 89 (1992) 21152119.
[48] J.J. Soghomonian, D.L. Martin, Two isoforms of glutamate decarboxylase: why?
Trends Pharmacol. Sci. 19 (1998) 500505.
[49] C.S. Pinal, A.J. Tobin, Uniqueness and redundancy in GABA production, Perspect.
Dev. Neurobiol. 5 (1998) 109118.
[50] I. Kass, D.E. Hoke, M.G. Costa, C.F. Reboul, B.T. Porebski, N.P. Cowieson, H. Leh,
E. Pennacchietti, J. McCoey, O. Kleifeld, C. Borri Voltattorni, D. Langley, B. Roome,
I.R. Mackay, D. Christ, D. Perahia, M. Buckle, A. Paiardini, D. De Biase, A.M. Buckle,
Cofactor-dependent conformational heterogeneity of GAD65 and its role in auto-
immunity and neurotransmitter homeostasis, Proc. Natl. Acad. Sci. U. S. A. 111
(2014) E2524E2529.
[51] G. Giardina, R. Montioli, S. Gianni, B. Cellini, A. Paiardini, C.B. Voltattorni,
F. Cutruzzola, Open conformation of human DOPA decarboxylase reveals the me-
chanism of PLP addition to Group II decarboxylases, Proc. Natl. Acad. Sci. U. S. A.
108 (2011) 2051420519.
[52] C. Lieberwirth, Y. Pan, Y. Liu, Z. Zhang, Z. Wang, Hippocampal adult neurogenesis:
its regulation and potential role in spatial learning and memory, Brain Res. 1644
(2016) 127140.
[53] S. Ge, D.A. Pradhan, G.L. Ming, H. Song, GABA sets the tempo for activity-depen-
dent adult neurogenesis, Trends Neurosci. 30 (2007) 18.
[54] F.E. Harrison, A.H. Hosseini, M.P. McDonald, Endogenous anxiety and stress re-
sponses in water maze and Barnes maze spatial memory tasks, Behav. Brain Res.
198 (2009) 247251.
[55] U. Rudolph, H. Mohler, Analysis of GABAA receptor function and dissection of the
pharmacology of benzodiazepines and general anesthetics through mouse genetics,
Annu. Rev. Pharmacol. Toxicol. 44 (2004) 475498.
[56] J. Declercq, B. Brouwers, V.P. Pruniau, P. Stijnen, G. de Faudeur, K. Tuand,
S. Meulemans, L. Serneels, A. Schraenen, F. Schuit, J.W. Creemers, Metabolic and
behavioural phenotypes in Nestin-Cre mice are caused by hypothalamic expression
of human growth hormone, PLoS One 10 (2015) e0135502.
[57] S. Li, T.P. Kumar, S. Joshee, T. Kirschstein, S. Subburaju, J.S. Khalili, J. Kloepper,
C. Du, A. Elkhal, G. Szabo, R.K. Jain, R. Kohling, A. Vasudevan, Endothelial cell-
derived GABA signaling modulates neuronal migration and postnatal behavior, Cell
Res. 28 (2018) 221248.
[58] U. Rudolph, H. Mohler, GABAA receptor subtypes: therapeutic potential in Down
syndrome, aective disorders, schizophrenia, and autism, Annu. Rev. Pharmacol.
Toxicol. 54 (2014) 483507.
[59] O. Marin, Interneuron dysfunction in psychiatric disorders, Nat. Rev. Neurosci. 13
(2012) 107120.
[60] B. Luscher, Q. Shen, N. Sahir, The GABAergic decit hypothesis of major depressive
disorder, Mol. Psychiatry 16 (2011) 383406.
[61] J.A. McQuail, C.J. Frazier, J.L. Bizon, Molecular aspects of age-related cognitive
decline: the role of GABA signaling, Trends Mol. Med. 21 (2015) 450460.
[62] A. Rozycka, M. Liguz-Lecznar, The space where aging acts: focus on the GABAergic
synapse, Aging Cell 16 (2017) 634643.
[63] K. Jannusch, C. Jockwitz, H.J. Bidmon, S. Moebus, K. Amunts, S. Caspers, A com-
plex interplay of vitamin B1 and B6 metabolism with cognition, brain structure, and
functional connectivity in older adults, Front. Neurosci. 11 (2017) 596.
[64] R.R. Schur, L.W. Draisma, J.P. Wijnen, M.P. Boks, M.G. Koevoets, M. Joels,
D.W. Klomp, R.S. Kahn, C.H. Vinkers, Brain GABA levels across psychiatric dis-
orders: a systematic literature review and meta-analysis of (1) H-MRS studies, Hum.
Brain Mapp. 37 (2016) 33373352.
[65] H. Mohler, The GABA system in anxiety and depression and its therapeutic po-
tential, Neuropharmacology 62 (2012) 4253.
[66] K.S. Smith, U. Rudolph, Anxiety and depression: mouse genetics and pharmacolo-
gical approaches to the role of GABA(A) receptor subtypes, Neuropharmacology 62
(2012) 5462.
[67] M. Bialer, H.S. White, Key factors in the discovery and development of new anti-
epileptic drugs, Nat. Rev. Drug Discov. 9 (2010) 6882.
[68] B. Lippert, B.W. Metcalf, M.J. Jung, P. Casara, 4-Amino-hex-5-enoic acid, a selective
catalytic inhibitor of 4-aminobutyric-acid aminotransferase in mammalian brain,
Eur. J. Biochem. 74 (1977) 441445.
[69] W.D. Shields, J.M. Pellock, Vigabatrin 35 years later - from mechanism of action to
benet-risk considerations, Acta Neurol. Scand. Suppl. (2011) 14.
[70] E. Ben-Menachem, Mechanism of action of vigabatrin: correcting misperceptions,
Acta Neurol. Scand. Suppl. (2011) 515.
[71] A. Represa, Y. Ben-Ari, Trophic actions of GABA on neuronal development, Trends
Neurosci. 28 (2005) 278283.
[72] W.C. Oh, S. Lutzu, P.E. Castillo, H.B. Kwon, De novo synaptogenesis induced by
GABA in the developing mouse cortex, Science 353 (2016) 10371040.
[73] A.A. Salari, A. Bakhtiari, J.R. Homberg, Activation of GABA-A receptors during
postnatal brain development increases anxiety- and depression-related behaviors in
a time- and dose-dependent manner in adult mice, Eur. Neuropsychopharmacol. 25
(2015) 12601274.
[74] S.J. Shabel, C.D. Proulx, J. Piriz, R. Malinow, Mood regulation. GABA/glutamate
co-release controls habenula output and is modied by antidepressant treatment,
Science 345 (2014) 14941498.
[75] M. Farrant, Z. Nusser, Variations on an inhibitory theme: phasic and tonic activa-
tion of GABA(A) receptors, Nat. Rev. Neurosci. 6 (2005) 215229.
[76] F. Crestani, K. L ow, R. Keist, M. Mandelli, H. Mohler , U. Rudolph, Mol ecular
targets for the my orelaxant acti on of diazepam, Mol. Pharmacol . 59 (2001)
442445.
[77] J.N. Crawley, J.K. Belknap, A. Collins, J.C. Crabbe, W. Frankel, N. Henderson,
R.J. Hitzemann, S.C. Maxson, L.L. Miner, A.J. Silva, J.M. Wehner, A. Wynshaw-
Boris, R. Paylor, Behavioral phenotypes of inbred mouse strains: implications and
recommendations for molecular studies, Psychopharmacology 132 (1997) 107124.
[78] K.R. Bailey, N.R. Rustay, J.N. Crawley, Behavioral phenotyping of transgenic and
knockout mice: practical concerns and potential pitfalls, ILAR J. 47 (2006)
124131.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
12
[79] I.Schneider,W.S.Tirsch,T.Faus-Kessler,L.Becker,E.Kling,R.L.Busse,A.Bender,
B. Feddersen, J. Tritschler, H. Fuchs, V. Gailus-Durner, K.H. Englmeier, M.H. de Angelis,
T. Klopstock, Systematic, standardized and comprehensive neurological phenotyping of
inbred mice strains in the German Mouse Clinic, J. Neurosci. Methods 157 (2006) 8290.
[80] S.C. Almo, J.B. Bonanno, J.M. Sauder, S. Emtage, T.P. Dilorenzo, V. Malashkevich,
S.R. Wasserman, S. Swaminathan, S. Eswaramoorthy, R. Agarwal, D. Kumaran,
M. Madegowda, S. Ragumani, Y. Patskovsky, J. Alvarado, U.A. Ramagopal,
J. Faber-Barata, M.R. Chance, A. Sali, A. Fiser, Z.Y. Zhang, D.S. Lawrence,
S.K. Burley, Structural genomics of protein phosphatases, J. Struct. Funct. Genom. 8
(2007) 121140.
[81] A. Seifried, J. Schultz, A. Gohla, Human HAD phosphatases: structure, mechanism,
and roles in health and disease, FEBS J. 280 (2013) 549571.
[82] A.M. Burroughs, K.N. Allen, D. Dunaway-Mariano, L. Aravind, Evolutionary geno-
mics of the HAD superfamily: understanding the structural adaptations and cata-
lytic diversity in a superfamily of phosphoesterases and allied enzymes, J. Mol. Biol.
361 (2006) 10031034.
E. Jeanclos et al. BBA - Molecular Basis of Disease xxx (xxxx) xxx–xxx
13
... These will be useful experimental tools to directly study whether increasing the levels of bioactive vitamin B6 in the brain may help those with mental health conditions associated with impaired memory, learning and mood. the cells are still largely unknown, it is clear that the intracellular availability of PLP for co-enzymatic functions depends on PLP carriers/scavengers and on the hydrolytic activity of pyridoxal 5'-phosphate phosphatase (PDXP) (Wilson et al., 2019;Ciapaite et al., 2023;Fux and Sieber, 2020;Jang et al., 2003;Jeanclos et al., 2019). ...
... We have previously shown that the genetic knockout of PDXP (PDXP-KO) in mice increases brain PLP levels and improves spatial memory and learning, suggesting that elevated PLP levels can improve cognitive functions in this model (Jeanclos et al., 2019). We therefore reasoned that a pharmacological inhibition of PDXP may be leveraged to increase intracellular PLP levels and conducted a highthroughput screening campaign to identify small-molecule PDXP modulators. ...
... Floxed PDXP mice (Pdxp tm1Goh ) were generated on a C57Bl/6J background, and whole-body Pdxp knockouts were achieved by breeding with B6.FVB-Tg(EIIa-cre)C5379Lmgd/J (EIIa-Cre) transgenic mice, as described (Jeanclos et al., 2019). All experiments were authorized by the local veterinary authority and committee on the ethics of animal experiments (Regierung von Unterfranken). ...
Article
Full-text available
Vitamin B6 deficiency has been linked to cognitive impairment in human brain disorders for decades. Still, the molecular mechanisms linking vitamin B6 to these pathologies remain poorly understood, and whether vitamin B6 supplementation improves cognition is unclear as well. Pyridoxal 5’-phosphate phosphatase (PDXP), an enzyme that controls levels of pyridoxal 5’-phosphate (PLP), the co-enzymatically active form of vitamin B6, may represent an alternative therapeutic entry point into vitamin B6-associated pathologies. However, pharmacological PDXP inhibitors to test this concept are lacking. We now identify a PDXP and age-dependent decline of PLP levels in the murine hippocampus that provides a rationale for the development of PDXP inhibitors. Using a combination of small-molecule screening, protein crystallography, and biolayer interferometry, we discover, visualize, and analyze 7,8-dihydroxyflavone (7,8-DHF) as a direct and potent PDXP inhibitor. 7,8-DHF binds and reversibly inhibits PDXP with low micromolar affinity and sub-micromolar potency. In mouse hippocampal neurons, 7,8-DHF increases PLP in a PDXP-dependent manner. These findings validate PDXP as a druggable target. Of note, 7,8-DHF is a well-studied molecule in brain disorder models, although its mechanism of action is actively debated. Our discovery of 7,8-DHF as a PDXP inhibitor offers novel mechanistic insights into the controversy surrounding 7,8-DHF-mediated effects in the brain.
... Further, studies in humans have delineated the function of a newly discovered PLP binding protein (PLPBP), which binds cellular PLP, acting as a regulator of PLP homeostasis intracellularly (Darin et al., 2016;Johnstone et al., 2019;Plecko et al., 2017). Both PL reductase and PLPBP are suggested to prevent adverse reactions of the aldehyde moiety of PL/PLP with non-specific cellular amino acids and amines (Johnstone et al., 2019;Ramos et al., 2019). ...
... organs (brain, muscle, red blood cells) and γ-aminobutyric acid in the brain, leading to phenotypical changes in behavior and cognition as well as decreased performance in motor tests (Jeanclos et al., 2019). This suggests that as-of-yet unidentified variants of PDXP could result in neurological disorders. ...
Article
Full-text available
To support EFSA in the preparatory work for the assessment of Tolerable Upper Intake Levels (UL) for vitamin B6, a tailored ‘high level’ protocol was developed, based on a template protocol and taking into account the specificities of vitamin B6. This protocol outlined the methods, the relevant endpoints and priority adverse health effects in relation to high intakes of vitamin B6. Systematic reviews (SR) were conducted following a tailored literature search, data extraction, evidence appraisal (i.e. risk of bias (RoB) assessment) and evidence synthesis. Narrative reviews (NR) were conducted to gather contextual evidence relevant to the interpretation of the main body of evidence. In the SR on the dose‐response relationship between vitamin B6 and peripheral neuropathy, 3793 records were identified and reduced to 32 individual records after screening for eligibility. The evidence appraisal revealed an overall moderate to high RoB of the individual studies and an overall rating of the total body of evidence as very low due mainly due to the type of data from case studies/case reports and limited number of other data. The available evidence confirmed a high degree of inter‐individual variability to sensitivity to a high exposure of vitamin B6 in relation to development of peripheral neuropathic outcomes but did not allow to determine a No Observed Adverse Effect Level or Lowest Observed Adverse Effect Level. In the SR on developmental toxicity, 4941 records were identified and reduced to 23 individual records including human and animal studies after screening for eligibility. The available data demonstrated a high degree of heterogeneity with respect to exposure and adverse health outcomes and overall, showed no positive or causal relationship between vitamin B6 intake and adverse developmental effects, including congenital defects. The NRs showed a paucity of data on high intake of vitamin B6 vitamers and their metabolism.
... The loss of PDXP has been shown to alter the balance between excitatory and inhibitory transmission at specific synapses in the body. For example, in the PDXP-KO mouse model, PLP levels increased threefold, GABA levels increased by 20%, and the mice showed mild anxiety-like behaviors [48], suggesting that increasing PLP levels increase GABA production. In addition, the level of L-serine and glycine were decreased in PDXP-KO mice [48]. ...
... For example, in the PDXP-KO mouse model, PLP levels increased threefold, GABA levels increased by 20%, and the mice showed mild anxiety-like behaviors [48], suggesting that increasing PLP levels increase GABA production. In addition, the level of L-serine and glycine were decreased in PDXP-KO mice [48]. Our results showed that PDXK expression was down-regulated, and the concentrations of L-serine and glycine were increased in offspring mice after exposure to PY during pregnancy. ...
Article
Full-text available
Pyridaben (PY) is a widely used organochlorine acaricide, which can be detected in the peripheral blood of pregnant women. Available evidence suggests that PY has reproductive toxicity. However, it remains uncertain whether prenatal PY exposure impacts neurobehavioral development in offspring. Here, we administered PY to pregnant mice at a dose of 0.5 and 5 mg kg⁻¹ day⁻¹ via gavage and observed anxiety-like behaviors in PY offspring aged five weeks. We then integrated the metabolome and transcriptome of the offspring's brain to explore the underlying mechanism. Metabolome data indicated that the vitamin B6 metabolism pathway was significantly affected, and the pyridoxal 5′-phosphate (PLP) concentration and the active form of vitamin B6 was significantly reduced. Moreover, the transcriptome data showed that both PLP generation-related Pdxk and anxiety-related Gad1 were significantly down-regulated. Meanwhile, there was a decreasing trend in the concentration of GABA in the hippocampal DG region. Next, we supplemented PLP at a dose of 20 mg kg⁻¹ d⁻¹ to the PY offspring via intraperitoneal injection at three weeks. We found up-regulated expression of Pdxk and Gad1 and restored anxiety-like behaviors. This study suggests that prenatal exposure to PY can disrupt vitamin B6 metabolism, reduce the concentration of PLP, down-regulate the expression levels of Pdxk and Gad1, inhibit the production of GABA, and ultimately lead to anxiety-like behaviors in offspring.
... According to previous research, we speculate that the alleviation of adverse reactions may be because vitamin B6 drives the production of GABA in the brain by regulating glutamate decarboxylase [16] and reversing the de-inhibition of the central vestibule, thereby alleviating the adverse reactions caused by minocycline. Pyridoxal phosphate (PLP) is the active form of vitamin B6 in the human body. ...
Article
Full-text available
Background Vitamin B6 is an essential water-soluble vitamin for humans. It is often used to prevent a variety of neuropathies, relieve vomiting, and relieve symptoms such as hand and foot neuritis. Aim To evaluate whether vitamin B6 can alleviate the adverse reactions caused by the quadruple anti-Helicobacter pylori treatment regimen containing minocycline and metronidazole. Methods In this randomized controlled trial, 280 patients with H. pylori infection were randomly placed into one of two treatment groups—the conventional treatment group and the vitamin B6 supplement treatment group—for 2 weeks. The primary endpoint was the total incidence of adverse reactions up to 2 weeks after treatment initiation. The study was designed according to CONSORT Medicinal Interventions. And it was registered with Chinese Clinical Trial Registry under the number ChiCTR2100053833. Results In terms of efficacy, vitamin B6 does not affect the efficacy of conventional regimen. In the vitamin B6 supplement treatment group, the incidence of adverse reactions was 56.92%, which was significantly lower than the 74.62% observed in the conventional treatment group. In addition, the severity of adverse reactions was also significantly reduced. The proportion of moderate to severe central nervous system symptoms decreased from 58.7 to 14.63%. And, the proportion of moderate to severe gastrointestinal reactions decreased from 33.33 to 0%. We speculate that the mechanism of vitamin B6 of reducing adverse reaction may be related to the production of GABA in the brain. Conclusions Vitamin B6 can alleviate adverse reactions of the quadruple anti-H. pylori regimen containing minocycline and metronidazole.
... Showing an isolated effect of vitamin B6, intraperitoneal injections of pyridoxine (350mg/kg BW) twice a day for 3 weeks in young C57Bl/6J mice improved novel object recognition memory and significantly raised 5-HT concentrations and the protein expression of tyrosine hydroxylase (TH), the rate-limiting enzyme for catecholaminergic neurotransmitter synthesis, in the hippocampus [168]. Consistent with these findings, high plasma and brain PLP (active form of B6) levels achieved by deletion of PLP-degrading enzyme in PDXP-null mice increased GABA levels in the brain by~20% and markedly enhanced spatial memory and motor performance [169]. The important role of vitamin B6 in brain health has also been demonstrated in animals other than mice. ...
Article
Full-text available
Alzheimer’s disease (AD) is the most common type of dementia that affects millions of individuals worldwide. It is an irreversible neurodegenerative disorder that is characterized by memory loss, impaired learning and thinking, and difficulty in performing regular daily activities. Despite nearly two decades of collective efforts to develop novel medications that can prevent or halt the disease progression, we remain faced with only a few options with limited effectiveness. There has been a recent growth of interest in the role of nutrition in brain health as we begin to gain a better understanding of what and how nutrients affect hormonal and neural actions that not only can lead to typical cardiovascular or metabolic diseases but also an array of neurological and psychiatric disorders. Vitamins and minerals, also known as micronutrients, are elements that are indispensable for functions including nutrient metabolism, immune surveillance, cell development, neurotransmission, and antioxidant and anti-inflammatory properties. In this review, we provide an overview on some of the most common vitamins and minerals and discuss what current studies have revealed on the link between these essential micronutrients and cognitive performance or AD.
... The larger C1 and C2 caps limit the access to the catalytic core, so that these enzymes preferentially convert substrates of low molecular weight. (Burroughs et al., 2006, Allen andDunaway-Mariano, 2009) Until now, only two phosphatases capped in such a way have been identified to act on macromolecules: C1-capped Eya3 dephosphorylates the C-terminal tyrosyl residue of histone H2A.X (Krishnan et al., 2009) and C2-capped pyridoxal phosphatase (PDXP; also called chronophin, CIN) dephosphorylates pyridoxal 5'-phosphate (PLP, the active form of vitamin B6) (Jang et al., 2003, Kestler et al., 2014, Jeanclos et al., 2019 as well as a serine residue of the actin-binding protein cofilin (Gohla et al., 2005). ...
Thesis
Full-text available
Mammalian phoshoglycolate phosphatase (PGP, also known as AUM) belongs to the ubiquitous HAD superfamily of phosphatases. As several other members of HAD phosphatases, the Mg2+-dependent dephosphorylation is conducted via a nucleophilic attack from a conserved aspartate residue in the catalytic cleft. The protein structure of PGP could not yet be solved entirely. Only a hybrid consisting of the PGP cap and the PDXP core (pyridoxal phosphatase, closest enzyme paralog) was crystallizable so far. PGP is able to efficiently dephosphorylate 2-phosphoglycolate, 2-phospho-L-lactate, 4-phospho-D-erythronate, and glycerol-3-phosphate in vitro which makes them likely physiological substrates. The first three substrates can be derived from metabolic side reactions (during glycolysis) and inhibit key enzymes in glycolysis and pentose phosphate pathway, the latter is situated at the intersection between glycolysis and lipogenesis. 2-phosphoglycolate can also be released in the context of repair of oxidative DNA damage. The activity of purified PGP can be reversibly inhibited by oxidation - physiologically likely in association with epidermal growth factor (EGF) signal transduction. In fact, an association between persistently lacking PGP activity (via downregulation) and the presence of hyperphosphorylated proteins after EGF stimulation has been identified. Reversible oxidation and transient inactivation of PGP may be particularly important for short-term and feedback regulatory mechanisms (as part of the EGF signaling). Furthermore, cellular proliferation in PGP downregulated cells is constantly reduced. Whole-body PGP inactivation in mice is embryonically lethal. Despite the many well-known features and functions, the knowledge about PGP is still incomplete. In the present work the influence of reactive oxygen species (ROS) on PGP activity in cells und a possible connection between oxidative stress and the proliferation deficit of PGP downregulated cells was investigated. For the experiments, a spermatogonial cell line was used (due to the high PGP expression in testis). PGP activity can be reversibly inhibited in cellular lysates by H2O2 (as a ROS representative). Reversible oxidation could thus indeed be physiologically important. More oxidative DNA damage (by bleomycin) showed no PGP-dependent effects here. EGF stimulation (as an inducer of transient and well-controlled ROS production), low concentrations of menadione (as an oxidant) and N-acetylcysteine (as an antioxidant) were able to approximate the proliferation rate in PGP downregulated cells to that of control cells. The redox regulation of PGP could thus have an influence on cellular proliferation as a feedback mechanism - a mechanism that could not take place in PGP downregulated cells. However, the connections are probably even more complex and cannot be elucidated by a sole examination of the proliferation rate. The present results can thus only be regarded as preliminary experiments. For a better understanding of the features and functions of PGP, this work then focused on specific regulation of enzyme activity by pharmacologically applicable small molecules. Four potent inhibitors had previously been identified in a screening campaign. In this work, three of these four inhibiting compounds could be further characterized in experiments with highly purified, recombinant murine and human PGP. Compounds #2 and #9 showed competitive inhibition properties with a markedly rising KM value with little or no change in vmax. The results were consistent for all tested protein variants: the murine and the human PGP as well as a PGP/PDXP hybrid protein. Compound #1 was the most potent and interesting PGP-inhibitory molecule: less change in KM and a constant decrease in vmax as well as a lower impact on the PGP/PDXP hybrid hint at a mixed mode of inhibition as a combination of competitive and non-competitive inhibition. The characterization of the potential inhibitors can serve as a basis for further structural analysis and studies on the complex physiological role of PGP.
... This led to improved spatial learning and memory but decreased motor performance and induced anxiety within the mice. 11 It has been shown that GABA plays a prominent role in movement disorders, but its ability to be modulated by the gene for pyridoxal phosphatase is a novel finding. 12 Several connections and contradictions to our case are noted in the literature. ...
Article
Full-text available
Tardive dyskinesia (TD) is a movement disorder associated with dopamine receptor blocking medications. Recommended treatments for TD include discontinuing the causative agent, adding vesicular monoamine transporter 2 (VMAT2) inhibitors, or adding vitamin B6. We present a 66-year-old Caucasian male with bipolar I disorder who developed TD while on lithium and quetiapine having been euthymic on this regimen for three years. He was initially treated with 1200 mg B6 daily. This failed to improve his TD and was associated with a depressive episode. He switched to valbenazine 40 mg daily which improved his TD and concurrently his mood, but months later the TD symptoms worsened again. Our case adds to the literature by demonstrating that some patients with TD will not respond to vitamin B6. To our knowledge, ours is the first case suggesting association of high dose vitamin B6 with depression. This case also demonstrates that response to valbenazine may not last and further studies are needed.
Preprint
Vitamin B6 deficiency has been linked to cognitive impairment in human brain disorders for decades. Still, the molecular mechanisms linking vitamin B6 to these pathologies remain poorly understood, and whether vitamin B6 supplementation improves cognition is unclear as well. Pyridoxal phosphatase (PDXP), an enzyme that controls levels of pyridoxal 5’-phosphate (PLP), the co-enzymatically active form of vitamin B6, may represent an alternative therapeutic entry point into vitamin B6-associated pathologies. However, pharmacological PDXP inhibitors to test this concept are lacking. We now identify a PDXP and age-dependent decline of PLP levels in the murine hippocampus that provides a rationale for the development of PDXP inhibitors. Using a combination of small molecule screening, protein crystallography and biolayer interferometry, we discover and analyze 7,8-dihydroxyflavone (7,8-DHF) as a direct and potent PDXP inhibitor. 7,8-DHF binds and reversibly inhibits PDXP with low micromolar affinity and sub-micromolar potency. In mouse hippocampal neurons, 7,8-DHF increases PLP in a PDXP-dependent manner. These findings validate PDXP as a druggable target. Of note, 7,8-DHF is a well-studied molecule in brain disorder models, although its mechanism of action is actively debated. Our discovery of 7,8-DHF as a PDXP inhibitor offers novel mechanistic insights into the controversy surrounding 7,8-DHF-mediated effects in the brain.
Preprint
Full-text available
Vitamin B6 deficiency has been linked to cognitive impairment in human brain disorders for decades. Still, the molecular mechanisms linking vitamin B6 to these pathologies remain poorly understood, and whether vitamin B6 supplementation improves cognition is unclear as well. Pyridoxal phosphatase (PDXP), an enzyme that controls levels of pyridoxal 5’-phosphate (PLP), the co-enzymatically active form of vitamin B6, may represent an alternative therapeutic entry point into vitamin B6-associated pathologies. However, pharmacological PDXP inhibitors to test this concept are lacking. We now identify a PDXP and age-dependent decline of PLP levels in the murine hippocampus that provides a rationale for the development of PDXP inhibitors. Using a combination of small molecule screening, protein crystallography and biolayer interferometry, we discover and analyze 7,8-dihydroxyflavone (7,8-DHF) as a direct and potent PDXP inhibitor. 7,8-DHF binds and reversibly inhibits PDXP with low micromolar affinity and sub-micromolar potency. In mouse hippocampal neurons, 7,8-DHF increases PLP in a PDXP-dependent manner. These findings validate PDXP as a druggable target. Of note, 7,8-DHF is a well-studied molecule in brain disorder models, although its mechanism of action is actively debated. Our discovery of 7,8-DHF as a PDXP inhibitor offers novel mechanistic insights into the controversy surrounding 7,8-DHF-mediated effects in the brain.
Article
Recently, biallelic variants in PLPBP coding for pyridoxal 5'-phosphate homeostasis protein (PLPHP) were identified as a novel cause of early-onset vitamin B6-dependent epilepsy. The molecular function and precise role of PLPHP in vitamin B6 metabolism are not well understood. To address these questions we used PLPHP deficient patient skin fibroblasts and HEK293 cells, and YBL036C (PLPHP ortholog) deficient yeast. We showed that independent of extracellular B6 vitamer type (pyridoxine, pyridoxamine or pyridoxal), intracellular PLP was lower in PLPHP deficient fibroblasts and HEK293 cells compared to controls. Culturing cells with pyridoxine or pyridoxamine led to the concentration-dependent accumulation of pyridoxine 5'-phosphate and pyridoxamine 5'-phosphate (PMP), respectively, suggesting insufficient pyridox(am)ine 5'-phosphate oxidase (PNPO) activity. Experiments utilizing 13C4-pyridoxine confirmed lower PNPO activity and revealed increased fractional turnovers of PLP and pyridoxal, indicating increased PLP hydrolysis to pyridoxal in PLPHP deficient cells. This effect could be partly counteracted by inactivation of pyridoxal phosphatase. PLPHP deficiency had a distinct effect on mitochondrial PLP and PMP, suggesting impaired activity of mitochondrial transaminases. Moreover, in YBL036C deficient yeast PLP was depleted and PMP accumulated only with carbon sources requiring mitochondrial metabolism. Lactate and pyruvate accumulation along with the decrease of tricarboxylic acid cycle intermediates downstream of α-ketoglutarate suggested impaired mitochondrial oxidative metabolism in PLPHP deficient HEK293 cells. We hypothesize that impaired activity of mitochondrial transaminases may contribute to this depletion. Taken together, our study provides new insights into the pathomechanisms of PLPBP deficiency and reinforces the link between PLPHP function, vitamin B6 metabolism and mitochondrial oxidative metabolism.
Article
Full-text available
The cerebral cortex is essential for integration and processing of information that is required for most behaviors. The exquisitely precise laminar organization of the cerebral cortex arises during embryonic development when neurons migrate successively from ventricular zones to coalesce into specific cortical layers. While radial glia act as guide rails for projection neuron migration, pre-formed vascular networks provide support and guidance cues for GABAergic interneuron migration. This study provides novel conceptual and mechanistic insights into this paradigm of vascular-neuronal interactions, revealing new mechanisms of GABA and its receptor-mediated signaling via embryonic forebrain endothelial cells. With the use of two new endothelial cell specific conditional mouse models of the GABA pathway (Gabrb3ΔTie2-Cre and VgatΔTie2-Cre), we show that partial or complete loss of GABA release from endothelial cells during embryogenesis results in vascular defects and impairs long-distance migration and positioning of cortical interneurons. The downstream effects of perturbed endothelial cell-derived GABA signaling are critical, leading to lasting changes to cortical circuits and persistent behavioral deficits. Furthermore, we illustrate new mechanisms of activation of GABA signaling in forebrain endothelial cells that promotes their migration, angiogenesis and acquisition of blood-brain barrier properties. Our findings uncover and elucidate a novel endothelial GABA signaling pathway in the CNS that is distinct from the classical neuronal GABA signaling pathway and shed new light on the etiology and pathophysiology of neuropsychiatric diseases, such as autism spectrum disorders, epilepsy, anxiety, depression and schizophrenia.
Article
Full-text available
Aging is associated with brain atrophy, functional brain network reorganization and decline of cognitive performance, albeit characterized by high interindividual variability. Among environmental influencing factors accounting for this variability, nutrition and particularly vitamin supply is thought to play an important role. While evidence exists that supplementation of vitamins B6 and B1 might be beneficial for cognition and brain structure, at least in deficient states and neurodegenerative diseases, little is known about this relation during healthy aging and in relation to reorganization of functional brain networks. We thus assessed the relation between blood levels of vitamins B1 and B6 and cognitive performance, cortical folding, and functional resting-state connectivity in a large sample of older adults (N > 600; age: 55–85 years), drawn from the population-based 1000BRAINS study. In addition to blood sampling, subjects underwent structural and functional resting-state neuroimaging as well as extensive neuropsychological testing in the domains of executive functions, (working) memory, attention, and language. Brain regions showing changes in the local gyrification index as calculated using FreeSurfer in relation to vitamin levels were used for subsequent seed-based resting-state functional connectivity analysis. For B6, a positive correlation with local cortical folding was found throughout the brain, while only slight changes in functional connectivity were observed. Contrarily, for B1, a negative correlation with cortical folding as well as problem solving and visuo-spatial working memory performance was found, which was accompanied by pronounced increases of interhemispheric and decreases of intrahemispheric functional connectivity. While the effects for B6 expand previous knowledge on beneficial effects of B6 supplementation on brain structure, they also showed that additional effects on cognition might not be recognizable in healthy older subjects with normal B6 blood levels. The cortical atrophy and pronounced functional reorganization associated with B1, contrarily, was more in line with the theory of a disturbed B1 metabolism in older adults, leading to B1 utilization deficits, and thus, an effective B1 deficiency in the brain, despite normal to high-normal blood levels.
Article
Full-text available
As it was established that aging is not associated with massive neuronal loss, as was believed in the mid-20th Century, scientific interest has addressed the influence of aging on particular neuronal subpopulations and their synaptic contacts, which constitute the substrate for neural plasticity. Inhibitory neurons represent the most complex and diverse group of neurons, showing distinct molecular and physiological characteristics and possessing a compelling ability to control the physiology of neural circuits. This review focuses on the aging of GABAergic neurons and synapses. Understanding how aging affects synapses of particular neuronal subpopulations may help explain the heterogeneity of aging-related effects. We reviewed the literature concerning the effects of aging on the numbers of GABAergic neurons and synapses as well as aging-related alterations in their presynaptic and postsynaptic components. Finally, we discussed the influence of those changes on the plasticity of the GABAergic system, highlighting our results concerning aging in mouse somatosensory cortex and linking them to plasticity impairments and brain disorders. We posit that aging-induced impairments of the GABAergic system lead to an inhibitory/excitatory imbalance, thereby decreasing neuron's ability to respond with plastic changes to environmental and cellular challenges, leaving the brain more vulnerable to cognitive decline and damage by synaptopathic diseases.
Article
Full-text available
Vitamin-B6-dependent epilepsies are a heterogenous group of treatable disorders due to mutations in several genes (ALDH7A1, PNPO, ALPL or ALDH4A1). In neonatal seizures, defects in ALDH7A1 and PNPO explain a major fraction of cases. Very recently biallelic mutations in PROSC were shown to be a novel cause in five families. We identified four further unrelated patients harbouring a total of six different mutations, including four novel disease mutations. Vitamin B6 plasma profiles on pyridoxine did not enable the differentiation of patients with PROSC mutations. All four patients were normocephalic and had normal cranial imaging. Pyridoxine monotherapy allowed complete seizure control in one, while two patients had occasional febrile or afebrile seizures and one needed additional valproate therapy for photosensitive seizures. Two patients underwent a controlled pyridoxine withdrawal with signs of encephalopathy within a couple of days. Three had favourable outcome with normal intellectual properties at age 12.5, 15.5 and 30 years, respectively, while one child had marked developmental delay at age 27 months. The clinical and electroencephalographic phenotype in patients with PROSC mutations was indistinguishable from ALDH7A1 and PNPO deficiency. We therefore confirm PROSC as a novel gene for vitamin-B6-dependent epilepsy and delineate a non-specific plasma vitamin B6 profile under pyridoxine treatment.
Article
Full-text available
Pyridoxal 5'-phosphate (PLP), the active form of vitamin B6, functions as a cofactor in humans for more than 140 enzymes, many of which are involved in neurotransmitter synthesis and degradation. A deficiency of PLP can present, therefore, as seizures and other symptoms that are treatable with PLP and/or pyridoxine. Deficiency of PLP in the brain can be caused by inborn errors affecting B6 vitamer metabolism or by inactivation of PLP, which can occur when compounds accumulate as a result of inborn errors of other pathways or when small molecules are ingested. Whole-exome sequencing of two children from a consanguineous family with pyridoxine-dependent epilepsy revealed a homozygous nonsense mutation in proline synthetase co-transcribed homolog (bacterial), PROSC, which encodes a PLP-binding protein of hitherto unknown function. Subsequent sequencing of 29 unrelated indivduals with pyridoxine-responsive epilepsy identified four additional children with biallelic PROSC mutations. Pre-treatment cerebrospinal fluid samples showed low PLP concentrations and evidence of reduced activity of PLP-dependent enzymes. However, cultured fibroblasts showed excessive PLP accumulation. An E. coli mutant lacking the PROSC homolog (Delta YggS) is pyridoxine sensitive; complementation with human PROSC restored growth whereas hPROSC encoding p.Leu175Pro, p.Arg241Gln, and p.Ser78Ter did not. PLP, a highly reactive aldehyde, poses a problem for cells, which is how to supply enough PLP for apoenzymes while maintaining free PLP concentrations low enough to avoid unwanted reactions with other important cellular nucleophiles. Although the mechanism involved is not fully understood, our studies suggest that PROSC is involved in intracellular homeostatic regulation of PLP, supplying this cofactor to apoenzymes while minimizing any toxic side reactions.
Article
Full-text available
Vitamin B6 includes six water-soluble vitamers: pyridoxal (PL), pyridoxamine (PM), pyridoxine (PN), and their phosphorylated forms. Pyridoxal 5'-phosphate (PLP) is an important cofactor for many metabolic enzymes. Several lines of evidence demonstrate that blood levels of PLP are significantly lower in patients with inflammation than in control subjects, and that vitamin B6 has anti-inflammatory effects, with therapeutic potential for a variety of inflammatory diseases. Although one of our group (NK) previously demonstrated that PL inhibits the NF-κB pathway, the molecular mechanism by which vitamin B6 suppresses inflammation is not well understood. Here, we showed that both PL and PLP suppressed the expression of cytokine genes in macrophages by inhibiting TLR-mediated TAK1 phosphorylation and the subsequent NF-κB and JNK activation. Furthermore, PL and PLP abolished NLRP3-dependent caspase-1 processing and the subsequent secretion of mature IL-1β and IL-18 in LPS-primed macrophages. In contrast, PM and PN had little effect on IL-1β production. PLP, but not PL, markedly reduced the production of mitochondrial reactive oxygen species (ROS) in peritoneal macrophages. Importantly, PL and PLP reduced the IL-1β production induced by LPS and ATP, or by LPS alone, in mice. Moreover, PL and PLP protected mice from lethal endotoxic shock. Collectively, these findings reveal novel anti-inflammatory activities for vitamin B6, and suggest its potential for preventing inflammatory diseases driven by the NLRP3 inflammasome.
Article
Heme oxygenase-1 (HO-1), which catalyzes heme degradation releasing iron, regulates several processes related to breast cancer. Iron metabolism deregulation is also connected with several tumor processes. However the regulatory relationship between HO-1 and iron proteins in breast cancer remains unclear. Using human breast cancer biopsies, we found that high HO-1 levels significantly correlated with low DMT1 levels. Contrariwise, high HO-1 levels significantly correlated with high ZIP14 and prohepcidin expression, as well as hemosiderin storage. At mRNA level, we found that high HO-1 expression significantly correlated with low DMT1 expression but high ZIP14, L-ferritin and hepcidin expression. In in vivo experiments in mice with genetic overexpression or pharmacological activation of HO-1, we detected the same expression pattern observed in human biopsies. In in vitro experiments, HO-1 activation induced changes in iron proteins expression leading to an increase of he-mosiderin, ROS levels, lipid peroxidation and a decrease of the growth rate. Such low growth rate induced by HO-1 activation was reversed when iron levels or ROS levels were reduced. Our findings demonstrate an important role of HO-1 on iron homeostasis in breast cancer. The changes in iron proteins expression when HO-1 is modulated led to the iron accumulation deregulating the iron cell cycle, and consequently, generating oxidative stress and low viability, all contributing to impair breast cancer progression.
Article
Anhedonia is the inability to experience pleasure from rewarding or enjoyable activities and is a core symptom of depression in humans. Here, we describe a protocol for the measurement of anhedonia in mice, in which anhedonia is measured by a sucrose preference test (SPT) based on a two-bottle choice paradigm. A reduction in the sucrose preference ratio in experimental relative to control mice is indicative of anhedonia. To date, inconsistent and variable results have been reported following the use of the SPT by different groups, probably due to the use of different protocols and equipment. In this protocol, we describe how to set up a clearly defined apparatus for SPT and provide a detailed protocol to ensure greater consistency when carrying out SPT. This optimized protocol is highly sensitive, reliable, and adaptable for evaluation of chronic stress-related anhedonia, as well as morphine-induced dependence. The whole SPT, including adaptation, baseline measurement, and testing, takes 8 d.
Article
Anxiety disorders result from a complex interplay of genetic and environmental factors such as stress. On the level of cellular signaling, regulator of G protein signaling 2 (Rgs2) has been implicated in human and rodent anxiety. However, there is limited knowledge about the role of Rgs2 in fear learning and reactivity to stress. In the present study, Rgs2(-/-) mice showed increased fear learning, male mice displayed increased contextual and cued fear learning, while females showed selectively enhanced cued fear learning. Male Rgs2(-/-) mice displayed increased long-term-contextual fear memory, but increased cued fear extinction. Learning in spatial non-aversive paradigms was also increased in Rgs2(-/-) mice. Female, but not male mice show increased spatial learning in the Barnes maze, while male mice showed enhanced place preference in the IntelliCage, rendering enhanced cognitive function non-specific for aversive stimuli. Consistent with previous results, Rgs2 deletion resulted in increased innate anxiety, including neophobic behavior expressed as hypo-locomotion, in three different tests based on the approach-avoidance conflict. Acute electric foot shock stress provoked hypo-locomotion in several exploration-based tests, suggesting fear generalization in both genotypes. Rgs2 deletion was associated with reduced monoaminergic neurotransmitter levels in the hippocampus and prefrontal cortex and disturbed corresponding GPCR expression of the adrenergic, serotonergic, dopaminergic and neuropeptide Y system. Taken together, Rgs2 deletion promotes improved cognitive function as well as increased anxiety-like behavior, but has no effect on acute stress reactivity. These effects may be related to the observed disruption of the monoaminergic systems.
Article
The active form of vitamin B6, pyridoxal 5'-phosphate (PLP), serves as a co-factor in more than 150 enzymatic reactions. Plasma PLP has consistently been shown to be low in inflammatory conditions; there is a parallel reduction in liver PLP, but minor changes in in erythrocyte and muscle PLP and in functional vitamin B6 biomarkers. Plasma PLP also predicts the risk of chronic diseases like cardiovascular disease and some cancers, and is inversely associated with numerous inflammatory markers in clinical and population-based studies. Vitamin B6 intake and supplementation improve some immune functions in vitamin B6-deficient humans and experimental animals. A possible mechanism involved is mobilization of vitamin B6 to the sites of inflammation where it may serve as a co-factor in pathways producing metabolites with immunomodulating effects. Relevant vitamin B6-dependent inflammatory pathways include vitamin B6 catabolism, the kynurenine pathway, sphingosine 1-phosphate metabolism, the transsulfuration pathway, and serine and glycine metabolism.