ArticlePDF Available

Abstract and Figures

We aimed to decipher the molecular genetic basis of disease in a cohort of children with a uniform clinical presentation of neonatal irritability, spastic or dystonic quadriplegia, virtually absent psychomotor development, axonal neuropathy, and elevated blood/CSF lactate. We performed whole-exome sequencing of blood DNA from the index patients. Detected compound heterozygous mutations were confirmed by Sanger sequencing. Structural predictions and a bacterial activity assay were performed to evaluate the functional consequences of the mutations. Mass spectrometry, Western blotting, and protein oxidation detection were used to analyze the effects of selenoprotein deficiency. Neuropathology indicated laminar necrosis and severe loss of myelin, with neuron loss and astrogliosis. In 3 families, we identified a missense (p.Thr325Ser) and a nonsense (p.Tyr429*) mutation in SEPSECS, encoding the O-phosphoseryl-tRNA:selenocysteinyl-tRNA synthase, which was previously associated with progressive cerebellocerebral atrophy. We show that the mutations do not completely abolish the activity of SEPSECS, but lead to decreased selenoprotein levels, with demonstrated increase in oxidative protein damage in the patient brain. These results extend the phenotypes caused by defective selenocysteine biosynthesis, and suggest SEPSECS as a candidate gene for progressive encephalopathies with lactate elevation. © 2015 American Academy of Neurology.
Content may be subject to copyright.
ARTICLES
Anna-Kaisa Anttonen,
MD, PhD*
Taru Hilander, MSc*
Tarja Linnankivi, MD,
PhD
Pirjo Isohanni, MD, PhD
Rachel L. French, BSc
Yuchen Liu, PhD
Miljan Simonovic, PhD
Dieter Söll, PhD
Mirja Somer, MD, PhD
Dorota Muth-Pawlak,
PhD
Garry L. Corthals, PhD
Anni Laari, MSc
Emil Ylikallio, MD, PhD
Marja Lähde, MD
Leena Valanne, MD, PhD
Tuula Lönnqvist, MD,
PhD
Helena Pihko, MD, PhD
Anders Paetau, MD, PhD
Anna-Elina Lehesjoki,
MD, PhD
Anu Suomalainen, MD,
PhD
Henna Tyynismaa, PhD
Correspondence to
Dr. Tyynismaa:
henna.tyynismaa@helsinki.fi
Supplemental data
at Neurology.org
Selenoprotein biosynthesis defect causes
progressive encephalopathy with elevated
lactate
ABSTRACT
Objective: We aimed to decipher the molecular genetic basis of disease in a cohort of children with
a uniform clinical presentation of neonatal irritability, spastic or dystonic quadriplegia, virtually
absent psychomotor development, axonal neuropathy, and elevated blood/CSF lactate.
Methods: We performed whole-exome sequencing of blood DNA from the index patients. De-
tected compound heterozygous mutations were confirmed by Sanger sequencing. Structural pre-
dictions and a bacterial activity assay were performed to evaluate the functional consequences of
the mutations. Mass spectrometry, Western blotting, and protein oxidation detection were used
to analyze the effects of selenoprotein deficiency.
Results: Neuropathology indicated laminar necrosis and severe loss of myelin, with neuron loss
and astrogliosis. In 3 families, we identified a missense (p.Thr325Ser) and a nonsense
(p.Tyr429*) mutation in SEPSECS, encoding the O-phosphoseryl-tRNA:selenocysteinyl-tRNA
synthase, which was previously associated with progressive cerebellocerebral atrophy. We show
that the mutations do not completely abolish the activity of SEPSECS, but lead to decreased
selenoprotein levels, with demonstrated increase in oxidative protein damage in the patient brain.
Conclusions: These results extend the phenotypes caused by defective selenocysteine biosynthe-
sis, and suggest SEPSECS as a candidate gene for progressive encephalopathies with lactate
elevation. Neurology
®
2015;85:110
GLOSSARY
PCH2D 5pontocerebellar hypoplasia type 2D; PEHO 5progressive encephalopathy with edema, hypsarrhythmia, and optic
atrophy; RC 5respiratory chain; SRM-MS 5selected reaction monitoringmass spectrometry; T
4
5thyroxine; tRNA 5
transfer RNA; TSH 5thyroid-stimulating hormone; T
3
5triiodothyronine.
Mitochondrial dysfunction is a frequent cause of childhood encephalopathy. Besides the typical
multisystemic disorders, an increasing number of mitochondrial defects are shown to cause a
CNS-specific phenotype.
15
Lactate elevation raises suspicion of mitochondrial involvement
and may be observed even in encephalopathies in which muscle biopsies show normal mito-
chondrial respiratory chain (RC) function.
13,6
Within our cohort of pediatric patients, we
identified patients with an undefined cause of cerebellocerebral atrophy, seizures, severe spas-
ticity, and axonal neuropathy with lactate elevation. We report that despite many of the clinical
and neuropathologic signs pointing toward mitochondrial impairment, the patients had
novel mutations in the SEPSECS gene, which functions in cytoplasmic transfer RNA
(tRNA)-charging in the selenoprotein biosynthesis pathway. We describe the uniform clinical,
neuroradiologic, and neuropathologic features of this entity and a detailed mutation
*These authors contributed equally to this work.
From the Department of Medical Genetics, Haartman Institute (A.-K.A., H.T.), Folkhälsan Institute of Genetics and Neuroscience Center (A.-K.A.,
A.L., A.-E.L.), Research Programs Unit, Molecular Neurology, Biomedicum Helsinki (T.H., P.I., A.L., E.Y., A.-E.L.), University of Helsinki;
Departments of Clinical Genetics (A.-K.A.) and Neurology (A.S.), Helsinki University Central Hospital; Department of Pediatric Neurology
(T. Linnankivi, P.I., T. Lönnqvist, H.P.), Childrens Hospital, University of Helsinki and Helsinki University Central Hospital, Finland;
Department of Biochemistry and Molecular Genetics (R.L.F., M. Simonovic), University of Illinois at Chicago; Department of Molecular Bio-
physics and Biochemistry (Y.L., D.S.), Yale University, New Haven, CT; Norio Centre (M. Somer), Department of Medical Genetics, Helsinki,
Finland; Turku Centre for Biotechnology (D.M.-P., G.L.C.), University of Turku and Åbo Akademi University; Department of Pediatric Neu-
rology (M.L.), South Karelia Central Hospital, Lappeenranta; Department of Radiology (L.V.), HUS Medical Imaging Center, Helsinki; and
Department of Pathology (A.P.), HUSLAB and University of Helsinki, Finland. G.L.C. is currently affiliated with Vant Hoff Institute for
Molecular Sciences, University of Amsterdam, the Netherlands.
Go to Neurology.org for full disclosures. Funding information and disclosures deemed relevant by the authors, if any, are provided at the end of the article.
© 2015 American Academy of Neurology 1
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
Published Ahead of Print on June 26, 2015 as 10.1212/WNL.0000000000001787
characterization. Moreover, our results indi-
cate oxidative damage in the brain as part of
the pathogenic mechanism resulting from se-
lenoprotein deficiency.
METHODS Standard protocol approvals, registrations,
and patient consents. All patient and control samples were
taken according to the Declaration of Helsinki, with informed
consent. The study was approved by the review board of the
Helsinki University Central Hospital.
The patients were identified within a cohort of 64 clinically
similar patients. One patient (patient 3) was part of the original
PEHO (progressive encephalopathy with edema, hypsarrhyth-
mia, and optic atrophy) syndrome patient series
7
and was
included in the neuroradiologic (group A) and ophthalmologic
study (patient 11) of that series.
A detailed neuropathologic examination was available for 3
patients (patients 1, 2, and 3), including the spinal cord from
patient 1; from patient 4, records pertaining to cerebellum, brain-
stem, and cerebral hemispheres were available. General autopsy
records were available from 3 patients (patients 1, 2, and 4).
Fresh-frozen tissue samples of patient 3 were available for the
study as well as fibroblasts of patients 1 and 2 and myoblasts of
patient 2.
DNA sequencing. For whole-exome sequencing, the exome
targets of the patientsDNA were captured with the
NimbleGen Sequence Capture 2.1M Human Exome v2.0 array
(NimbleGen, Basel, Switzerland) followed by sequencing with
the Illumina Genome Analyzer-IIx platform (Illumina, Inc.,
San Diego, CA) with 2 382 base pair paired-end reads. The
variant calling pipeline of the Finnish Institute for Molecular
Medicine was used for the reference genome alignment and
variant calling.
8
The coding exons of SEPSECS were sequenced
by Sanger sequencing.
Structural analysis of the mutations. Structural analysis was
based on the crystal structure of the human SEPSECS-tRNA
Sec
binary complex (PDB ID: 3HL2). The SEPSECS mutants
p.Thr325Ser and p.Tyr429*were generated in silico and analyzed
in Coot.
9
All figures were produced in PyMOL (The PyMOL
Molecular Graphics System, version 1.5.0.4, Schrödinger, LLC).
Oxyblot. The brain protein lysates were extracted using RIPA
buffer, and Oxyblot method was performed using an OxyBlot
Protein Oxidation Detection Kit (Millipore Corp., Billerica,
MA) according to the manufacturers instructions. The e-Meth-
ods on the Neurology
®
Web site at Neurology.org include full
descriptions of haplotype analysis, in vivo activity assay, and
protein analysis methods.
RESULTS Clinical data. We investigated 4 children
from 3 unrelated Finnish families. Clinical features
of the patients are summarized in table 1. These chil-
dren were born after uncomplicated pregnancies at
term to healthy nonconsanguineous parents. Two
patients were microcephalic at birth. The children
were irritable from birth and presented by the age
of 1 to 2 months with opisthotonus posturing, absent
head control, tremors, and myoclonic jerks. Severe
spastic or dystonic quadriplegia with absent psycho-
motor development became evident during the first
few months. Three patients had epileptic seizures,
including infantile spasms. As a sign of peripheral
neuropathy, the deep tendon reflexes attenuated or
vanished by the age of 2.5 years. The optic discs were
pale but not atrophic. All patients had edema of
hands, feet, and face, as well as narrow forehead,
tapering fingers, and high palate.
In 2 patients, early EEG studies (younger than 6
months of age) were normal, and later, hypsarrhythmia
with infantile spasms was documented. Later EEG re-
cordings showed severe slowing of background activity.
Sensory axonal neuropathy was verified by sural nerve
biopsy and electroneuromyography (table 1).
Two patients had elevated blood lactate levels and
one of them also had elevated lactate in the CSF. Pa-
tients 1 and 2 showed mild elevation of thyroid-
stimulating hormone (TSH) with normal levels of
thyroxine (T
4
) (table 1). Triiodothyronine (T
3
) levels
were not available. Other laboratory evaluations,
including liver transaminases, were unremarkable.
Neuroradiology and neuropathology showed neuron and
myelin loss. Neuroradiologic examinations showed
progressive cerebellar atrophy and less pronounced
cerebral atrophy (table e-1). Myelination was delayed
in the early MRIs and subsequently arrested. Cerebral
white matter showed pronounced and progressive
volume loss (figure 1).
Neuropathologic analysis revealed that all 4 pa-
tients shared features of progressive neuronal degener-
ation: laminar subtotal necrosis of the neocortex,
which was especially pronounced in the parietooccip-
ital regions (figure 2A), with relative sparing of the
hippocampi. The white matter showed myelin loss
and pallor with gliosis, consistent with degeneration
secondary to the neuronal loss. Patient 1 had subtotal
striatal degeneration (figure 2B) and also some tha-
lamic atrophy. All cases shared a quite severe degen-
eration and atrophy of the brainstem and cerebellar
cortex, creating an olivopontocerebellar atrophylike
appearance: basis pontis, inferior olives, and tegmen-
tum of the medulla oblongata were severely atrophic;
the cerebellar cortex was severely atrophic as well
(figure 2, C and D). Here, the molecular layer was
thin, accompanied by a subtotal loss of Purkinje cells
and a very thin granule cell layer (figure 2E). The
spinal cord, available from one case, showed atrophy
and degeneration especially in the posterior columns.
Finally, the general autopsy revealed mild to moder-
ate mostly microvacuolar fatty degeneration of the
liver parenchyma (figure 2F).
SEPSECS mutations identified as the genetic cause. The
genetic cause of the disease was identified by whole-
exome sequencing of DNA samples of 2 patients
(patients 1 and 2) from 2 unrelated families. The
identified variants were first filtered to exclude
nongenic variants and those common in populations.
2Neurology 85 July 28, 2015
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
On the basis that both families were of Finnish
ancestry and the clinical manifestation of both
patients closely resembled each other, we searched for
homozygous or compound heterozygous variants that
the patients shared. Two novel heterozygous variants
in SEPSECS were subsequently identified in both
patients, c.974C.G in exon 8 leading to a missense
mutation p.Thr325Ser and c.1287C.Ainexon11
leading to a nonsense mutation p.Tyr429*(RefSeq
NM_016955.3). The variants were validated by
Sanger sequencing (figure 3A). The parents of patient
1 and the mother of patient 2 were heterozygous
carriers; the DNA sample of the father of patient 2
was not available. The variants were not present in the
1000 Genomes database (www.1000genomes.org) or in
the NHLBI GO ESP Exome Variant Server (evs.gs.
washington.edu/EVS/) or in approximately 230
screened Finnish control samples. However, a recent
database of 3,323 exome sequences of Finnish
individuals provided a heterozygous carrier frequency
of 1:277 for the c.1287C.A, p.Tyr429*variant
(Sequencing Initiative Suomi, sisu.fimm.fi), suggesting
Table 1 Summary of clinical and laboratory examinations of patients
Study
Patient 1, family 1 Patient 2, family 2 Patient 3, family 3 Patient 4, family 3
Sex MFFF
Gestational age, wk 41 16 39 41 At term
Weight at birth, g 4,000 3,700 3,120 2,820
OFC at birth, cm (SD) 36.5 (11) 35.5 (10.5) 31 (23.3) 32 (22.3)
Presenting signs (age at
onset)
Irritability (neonatal),
opisthotonus (2 mo) Irritability, opisthotonus
(neonatal) Irritability, opisthotonus
(1.5 mo) NA
Tremors/myoclonus 111NA
Epileptic seizures (age at
onset)
Infantile spasms (12 mo) Infantile spasms (11 mo) 1NA
Spastic or dystonic
quadriplegia
1111
Central visual impairment 1111
Optic atrophy 222NA
Intellectual disability Profound Profound Profound Profound
Dysmorphic features
High arched palate 111NA
Tented upper lip 211NA
Outward turning ear
lobules
112NA
Narrow forehead 111NA
Edema of hands and feet 111NA
Tapered fingers 111NA
EEG (age) N (3 and 5 mo), hypsarrhythmia
(12 mo), very slow background
(6 y)
N (3 and 4 mo),
hypsarrhythmia (13 mo) Slow background (4 mo),
multifocal spikes, very slow
background (10 y)
Unspecific slowing
(14 mo)
Muscle biopsy N Variable fiber size,
neurogenic damage NNA
Sural nerve biopsy (age at
investigation)
Axonal neuropathy (2 y) Axonal neuropathy (2.3 y) Axonal neuropathy (10 y) NA
Neurography MCV low N, motor ampl N,
sensory ampl: radialis Y, suralis
(6 y)
MCV low N, Motor ampl N,
Sensory ampl: radialis Y,
suralis(2.3 y)
NA NA
Blood lactate (ref. 0.71.8
mmol/L)
4.9, 0.8, 5.6, 3.1 0.4, 2.8, 5.9, 4.2 N NA
CSF lactate (ref. 0.62.7
mmol/L)
1.5 1.4, 4.2 1.7 NA
Age at death, y 8.5 4.3 15.3 2.7
Abbreviations: ampl 5amplitudes; MCV 5motor nerve conduction velocity; N 5normal; NA 5not available; OFC 5occipital frontal circumference; ref. 5
reference.
Symbols: 25absent; 15present; Y5decreased.
Neurology 85 July 28, 2015 3
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
enrichment of the nonsense variant in Finland, but
without homozygous occurrence.
Next we screened 11 SEPSECS exons for mutations
in additional patients with mitochondrial encephalopa-
thy and/or other shared features. One patient (patient 3)
from an affected sib-pair with similar clinical findings
was found to be compound heterozygous for the
same SEPSECS mutations c.974C.G(p.Thr325Ser)
and c.1287C.A(p.Tyr429*). The DNA sample of
the affected sib was not available for the study. Inves-
tigation of the family histories of the 3 patients with
shared SEPSECS mutations revealed that they all
originated from a restricted area in eastern Finland.
Haplotype analysis of the nearby microsatellite markers
indicated shared ancestral haplotypes, further support-
ing the distant common origin of the mutations in our
patients (figure 3B).
Predicted effects of the mutations onto the SEPSECS
protein structure. SEPSECS codes for O-phosphoseryl-
tRNA:selenocysteinyl-tRNA synthase, the key en-
zyme in the sole biosynthetic route to selenocysteine
(Sec) in eukaryotes and archaea.
10,11
Residue Thr325,
which is affected by a missense mutation in our patients,
is a highly conserved amino acid in eukaryotic
SEPSECS (figure 3C). In the SEPSECS structure,
Thr325 is located in the middle of helix a12 in the
C-terminal domain of the protein (figure 3D). Helix
a12 provides support for the major elements that con-
stitute the active site of SEPSECS. Thr325 interacts
only with the backbone and side-chain atoms of helix
a12, and its replacement with serine may destabilize the
structure of a-helix. This could lead to altered
positioning of the cofactor pyridoxal phosphate, and
the floor (helix a10) and the clefts (loop 70 and
P-loop) of the active site. These structural
rearrangements in the catalytic pocket would
ultimately yield an enzyme with reduced catalytic
power.
The p.Tyr429*mutant messenger RNA escapes
nonsense-mediated decay, and is predicted to lead to
truncation of the 73 C-terminal amino acids of SEP-
SECS, the region critical both for tRNA binding and
enzyme activity (figure e-1). The full-length SEP-
SECS of approximately 55 kDa was readily detected
by Western blottingeven in a higher amount than
in the control sample (figure 3E)in the brain
autopsy sample of patient 3, but no evidence of a
truncated protein (;47 kDa) was found, indicating
that it was either not produced or rapidly degraded.
Figure 1 Brain imaging findings of SEPSECS deficiency
T2 and fluid-attenuated inversion recovery images of patient 1 at age 5 months (A) and 2 years, 10 months (B, C) and patient
2 at age 6 months (D) and 1 year, 8 months (E, F). The early images (A, D) show loss of periventricular white matter volume
and almost complete lack of myelin (arrows). Upon disease progression (B, C, E, F), widening of both central and cortical CSF
spaces and cerebellar atrophy are present, with no signal for myelin.
4Neurology 85 July 28, 2015
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
To functionally confirm the mutation effects, we
showed that both mutations severely affected
SEPSECSactivityinvivoinananaerobicEsche-
richia coli assay.
12
For the assay, we utilized the
DselA strain and inspected the ability of human
SEPSECStorestorethebenzylviologenreducing
activity of an Ecoliselenoprotein, the formate
dehydrogenase H. As predicted, p.Tyr429*mutant
was completely inactive in this assay, while
the p.Thr325Ser mutant was active albeit at the
decreased level (figure 3F).
Functional consequences of the SEPSECS mutations. In-
activating SEPSECS mutations presumably inhibit
synthesis of 25 selenoproteins (the human selenopro-
teome), which participate in diverse biological processes.
13
We measured selenoprotein levels in the lysate ob-
tained from the autopsy brain material of patient 3
using selected reaction monitoringmass spectro-
metry (SRM-MS). Protein levels were normalized
against glyceraldehyde 3-phosphate dehydrogenase
andphosphoglyceratekinase1.Tovalidatethe
method, the levels of the glial fibrillary acidic protein,
Figure 2 Histologic findings of SEPSECS deficiency
(A) Parietooccipital cortex displaying edemic transcortical laminar necrosis (ln) with a subtotal neuronal loss (arrows). (B)
Putamen (put) is severely neuron-depleted and gliotic (arrows). (C) Pontine basis (pb) is narrow, both neurons and transverse
fibers are reduced. (D) Atrophy of the inferior olives (io) and hili with olivocerebellar fibers and narrowed tegmentum seen in
low magnification of the medulla oblongata. (E) Atrophic cerebellar cortex, the molecular and granular layers are thin, with
practically total loss of Purkinje cells (arrows). (F) Liver parenchyma exhibiting a moderate microvacuolar fatty degeneration
(fat visible as white droplets). Paraffin sections, hematoxylin & eosin (A, C, D, F), Luxol fast bluecresyl violet (B, E); original
magnification 340 (A, E), 3100 (B), 310 (C, D), 3400 (F).
Neurology 85 July 28, 2015 5
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
which resides in astrocytes, were measured and shown
to be increased, thus indicating astrogliosis (figure 4,
A, D). In contrast, levels of myelin basic protein
and neurofilament medium were clearly reduced
(figure 4A), which is consistent with the observed
myelin and neuron loss in the patients. Three
selenoproteins, thioredoxin reductase TXNRD1 and
glutathione peroxidases GPX1 and GPX4, were
Figure 3 SEPSECS gene defect with structural and functional consequences of the mutations
(A) SEPSECS mutation sequences. The arrow indicates th em utation site. (B) Haplotypes on the chromosome 4 region containing
the SEPSECS gene in 3 patients from 3 unrelated families. Parent samples were used to construct the haplotypes when
available. (C) Alignment of the sequence region containing Thr325 (ar row) in eukaryo tic SEPSECS pr oteins. (D) Thr 325 is located
in the middle of helix a12 in the C-terminal domain of the protein. Helix a12 provides support for the major elements that
constitute the active site of SEPSECS by 2 direct interactions: (1) helix a12 (orange) interacts with helix a10 (from monomer 1 ,
in gray); (2) helix a10 carries a catalytic residue Lys284 (orange sticks) to which the cofactor PLP (orange sticks) is covalently
attached and forms the floor of the active site. The a10a12 interaction places Thr325 (orange sticks) behind the active-site
pocket and approximately 15 Å away from PLP (shown with up-and-down arrow). In addition, helix a12 interacts with helix a3
(blue) from the second SEPSECS monomer (monomer 2, in light blue), which is flanked by loop 70 and P-loop (blue) that form the
cleftsoftheactivesite.TheCCAendoftRNA
Sec
(beige) binds in the proximity of the P-loop, which is also implicated in binding of
the phosphoseryl group. (E) Sodium dodecyl sulfatepolyacrylamide gel electrophoresis from brain samples; SEPSECS patient
(P) and control (C). The full-length SEPSECS of approximately 55 kDa was readily detected, but no signal of the size of the
truncated protein (;47 kDa)was present.(F) In vivo dilution series assay of the ability of humanSEPSECS variants to restore the
benzyl viologenreducing activity of the selenoprotein formate dehydrogenase H in the Escherichia coli DselA deletion strain.
GAPDH 5glyceraldehyde 3-phosphate dehydrogenase; PLP 5pyridoxal phosphate; tRNA 5transfer RNA.
6Neurology 85 July 28, 2015
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
abundant enough to be reliably detected. Their levels
were decreased by 15% to 40% in the patient brain
sample compared with controls (figure 4B). Consistent
with a partial defect in selenoprotein production, levels
of TXNRD1 and TXNRD2 were decreased in the
patients brain as shown by Western blotting (figure
4D). However, the steady-state level of tRNA
Sec
was
not altered (figure 4F). Of note, the observed defects
were tissue-specific, as the patients fibroblasts,
myoblasts, or differentiated myotubes did not show
reduced TXNRD levels (data not shown).
Because of the lactate elevation, we analyzed the
amounts of mitochondrial RC complexes IIV in
the autopsy brain samples of patient 3 by blue native
electrophoresis, but found them to be similar to con-
trols (figure 4E). In addition, when we quantified the
amounts of RC complex subunits by SRM-MS, they
were shown to be mostly unaffected (figure 4C).
Figure 4 Selenoprotein and respiratory chain protein amounts in SEPSECS deficiency
(AC) Selected reaction monitoringmass spectrometry; autopsy brain sample of patient 3 and 3 controls. The results are
shown for (A) glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), and neurofilament medium (NFM), markers of
brain cells, for (B) selenoproteins glutathione peroxidases 1 and 4 (GPX1 and GPX4) and thioredoxin reductase 1 (TRXR1)
and for (C) mitochondrial respiratory chain complex subunits: NADH dehydrogenase (ubiquinone) Fe-S protein 1 (NDUFS1),
NADH dehydrogenase (ubiquinone) Fe-S protein 8 (NDUFS8), ubiquinol-cytochrome creductase core protein II (QCR2),
cytochrome coxidase subunit Va (COX5A), mitochondrially encoded ATP synthase 6 (ATPA), and citrate synthase (CISY).
(D) Western blotting of the brain samples for GFAP and 2 selenoproteins (P, patient 3; C, controls 13). (E) Blue native
electrophoresis of mitochondrial respiratory chain complexes in the patient (P) and control (C) brain samples. (F) Steady-
state levels of tRNA
Sec
compared with mitochondrial tRNA
Ala
in the brain sample of patient 3 (P) and controls (C1C4). (G)
Oxyblot shows increased amounts of oxidized proteins in patient brain (P) compared with controls (C1C3). GAPDH 5
glyceraldehyde 3-phosphate dehydrogenase; mt-tRNA 5mitochondrial transfer RNA; tRNA
Sec
5selenocysteine-specific
transfer RNA.
Neurology 85 July 28, 2015 7
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
Because the decreased selenoprotein synthesis
affected glutathione peroxidases and thioredoxin
reductasesenzymes of antioxidant defensewe
analyzed protein carbonylation in the patients
brain, and found protein oxidation to be clearly
increased (figure 4G).
DISCUSSION We report here that pathogenic muta-
tions in SEPSECS lead to severe cerebellocerebral
atrophy by attenuating the synthesis of selenopro-
teins, which leads to considerable oxidative damage
in the patient brains.
SEPSECS mutations were previously described in
a single report, underlying progressive cerebellocere-
bral atrophy with profound mental retardation, pro-
gressive microcephaly, severe spasticity, and
myoclonic or generalized tonic-clonic seizures, later
classified as pontocerebellar hypoplasia type 2D
(PCH2D) (MIM 613811).
12,14
The MRI findings
of progressive cerebellar atrophy, followed by cerebral
atrophy involving both white and gray matter, mim-
icked the findings in our patients. In contrast to a
normal metabolic profile of patients with PCH2D,
12
our patients had lactacidemia, and they also presented
with axonal neuropathy. Similarities between the pa-
tients with PCH2D and the patients described here
support the disease-causing role of the identified mu-
tations, thus extending the phenotypes caused by
defective selenocysteine biosynthesis.
The neuropathologic changes of SEPSECS defi-
ciency, such as progressive neuronal degeneration,
more pronounced laterally than midline, are remi-
niscent of Alpers syndrome,
15
which is caused by
defects in mitochondrial proteins, most commonly
in polymerase gamma, but also in Twinkle helicase
and the phenylalanyl-tRNA synthetase.
4,16,17
The
topography of the lesions corresponds to highly
energy-dependent regions of the CNS; in case of
SEPSECS deficiency, this is particularly the parieto-
occipital region. The patients with SEPSECS muta-
tions described here also showed moderate
degeneration of the liver postmortem. These find-
ings, together with elevated lactate, suggest that
encephalopathy due to SEPSECS deficiency and
mitochondrial encephalopathies, such as Alpers syn-
drome, could share some common pathogenic
mechanisms. However, we did not identify signifi-
cant alterations in the mitochondrial RC complexes
in the patients brain sample that would explain
changes typical for RC deficiencies.
Similar manifestations between mitochondrial and
selenoprotein disorders could be explained by the role
selenoproteins have in maintaining the cellular redox
potential and H
2
O
2
detoxification.
13
In other words,
selenoproteins are an important part of the antioxi-
dant defense. We showed remarkable increase of
protein carbonylation as a sign of oxidative stress in
the brain of patients with SEPSECS deficiency. As
mitochondria are one of the main sources of cellular
reactive oxygen species, SEPSECS deficiency could
especially damage cells with high mitochondrial activ-
ity. Future work on mouse models is needed to clarify
this aspect of selenoprotein pathology.
Progressive cerebellar atrophy of our patients
together with infantile spasms, dysmorphic features,
and edema of hands, feet, and face resembled the
findings typically seen in PEHO syndrome.
7,18
How-
ever, unlike patients with PEHO, our patients did
not have optic atrophy and were spastic rather than
hypotonic. Previously, a connection between ponto-
cerebellar hypoplasias, mitochondrial encephalopa-
thies, and PEHO-like features has been proposed in
PCH6 that is caused by RARS2 mutations.
19
RARS2
encodes the mitochondrial aminoacyl-tRNA synthe-
tase essential for charging tRNA
Arg
for protein synthe-
sis of mitochondrial RC complexes.
6
Of note,
SEPSECS also affects cellular functions through
tRNA, but in a different biological pathway, by cat-
alyzing the conversion of the phosphoseryl-tRNA
Sec
intermediate into selenocysteinyl-tRNA
Sec
.
10,11
Fur-
thermore, PCH2 types A, B, and C are caused by
mutations in genes encoding for subunits of the
tRNA-splicing endonuclease complex, TSEN54,
TSEN2 and TSEN34, respectively.
20
This complex
performs the splicing of intron-containing tRNAs,
which comprise approximately 6% of human tRNAs
needed for cytoplasmic translation.
21
Whether a com-
mon mechanism exists by which the defects of the
different cellular protein synthesis processes lead to
similar neurodegenerative phenotypes or whether
tRNA involvement in all these entities is purely coin-
cidental remains to be established.
SEPSECS is a tetramer composed of 2 dimers,
where each dimer contains 2 active sites formed at
the dimer interface.
10,11
The novel compound heter-
ozygous mutations identified in this study were pre-
dicted to have differential effects on the function of
the tetrameric SEPSECS: p.Thr325Ser introduced
Ser in the middle of an a-helix predicting destabili-
zation of the helix,
22
thereby modifying the catalytic
pocket of the enzyme, whereas p.Tyr429*resulted in
a total loss of function. Our dilution series of the
bacterial in vivo assay verified these predictions.
The previously reported SEPSECS mutations of pa-
tients with PCH2D displayed no detectable activity
in an anaerobic in vivo assay for SEPSECS activity.
12
Thus, the differences in the phenotypes of our pa-
tients and those reported previously may be caused by
differences in the residual SEPSECS activity. The fact
that the fibroblasts, myoblasts, and myotubes of our
patients did not display decreased selenoprotein levels
(not shown) suggests that the residual SEPSECS
8Neurology 85 July 28, 2015
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
activity was sufficient to maintain selenoprotein syn-
thesis in these cell types but not in the brain.
The 25 human selenoproteins function in remark-
ably diverse processes. Besides SEPSECS deficiency,
the only known human disease affecting selenopro-
tein synthesis is caused by mutations in SECISBP2,
which encodes a protein that recognizes the specific
insertion sequence in the selenoprotein messenger
RNAs. SECISBP2 mutations cause either a multisys-
tem disorder
23
or abnormal thyroid hormone metab-
olism with elevated TSH, T
4
, and reverse T
3
and
reduced T
3
levels.
24
Our patients presented with
increased TSH and normal T
4
levels, as measured
at ages 2 and 6 years. Thus, although affecting the
common metabolic pathway, mutations in SE-
CISBP2 and SEPSECS yield distinct phenotypes.
The selenoproteome is essential for mammals as
shown by the early embryonic lethality of the tRNA
Sec
gene (Trsp) knockout mouse.
25
The selenoprotein
knockout mice have further clarified their importance
for brain function.
26
The neuron-specific knockout
mice of GPx4,
27
Txnrd1,
28
and Trsp
29
are characterized
by neurodegeneration, in general, and by cerebellar
hypoplasia, in particular.
30
In the Txnrd1 mice, the
effect was attributed to decreased proliferation of gran-
ule cell precursors within the external granular layer.
28
Furthermore, the full knockout of SelP,
31
which enco-
des a plasma protein SELP that transports selenium
from the liver to peripheral tissues, was shown to have
a neurologic phenotype with spasticity and seizures.
32
It
is likely that the lack of several selenoproteins contrib-
utes to the neuronal loss in the selenoprotein biosyn-
thesis defects. Our SRM-MS and Western blotting
results derived from the analysis of an autopsy brain
sample from a patient with SEPSECS mutations
showed that selenoproteins were present albeit at sig-
nificantly reduced levels. However, because of the
severe neuronal loss and astrogliosis in the patient brain,
a comparative analysis has limitations. For instance,
neurons may have had more severe reductions of sele-
noproteins before their death. Also, the measurement of
selenoenzyme activities was not feasible with this mate-
rial. Regardless of these shortcomings, our result of
SEPSECS mutations causing a selenoprotein deficiency
in human brain implicates a specific requirement of
selenoproteins in postnatal brain development.
Based on the results of our study, we suggest SEP-
SECS sequencing in progressive early childhood brain
atrophies of unknown cause, especially when patients
present with sensory axonal neuropathy and elevated
lactate.
AUTHOR CONTRIBUTIONS
A.-K.A. collected and analyzed patient data and drafted the manuscript.
T.H., A.L., and E.Y. sequenced patients and performed protein and cell
culture experiments. T. Linnankivi and P.I. contributed to the clinical
analysis. R.L.F. and M. Simonovic performed the structural analysis.
Y.L. and D.S. performed the in vivo bacterial assay. D.M.-P. and
G.L.C. performed the mass spectrometry analysis. M. Somer, M.L.,
T. Lönnqvist, and H.P. contributed to the clinical analysis. L.V. per-
formed neuroradiology. A.P. performed neuropathology. H.T. performed
the exome sequencing analysis. A.-E.L., A.S., and H.T. designed the
study and drafted the manuscript. All authors revised the manuscript.
ACKNOWLEDGMENT
Anu Harju and Riitta Lehtinen are thanked for technical help. The authors
acknowledge the exome capture, sequencing, and variant calling pipeline
analysis performed by the Institute for Molecular Medicine Finland
FIMM, Technology Centre, and University of Helsinki. Proteome analysis
was made possible through use of the National Proteomics and Metabolo-
mics infrastructure of Biocenter Finland.
STUDY FUNDING
The authors thank the Sigrid Jusélius Foundation, Academy of Finland
and University of Helsinki (to H.T. and A.S.), Jane and Aatos Erkko
Foundation (to A.S.), Folkhälsan Research Foundation (to A.-E.L.), the
US NIH grant GM097042 (to M. Simonovic), the US National Institute
of General Medical Sciences GM22854 (to D.S.), Arvo and Lea Ylppö
Foundation (to A.-K.A. and H.T.), Orion Farmos Research Foundation
(to H.T.), Helsinki University Central Hospital Research Fund (to A.-K.
A.), and Foundation for Pediatric Research (to P.I.) for funding support.
DISCLOSURE
A. Anttonen has received research support from Arvo and Lea Ylppö
Foundation and Helsinki University Central Hospital Research Fund.
T. Hilander and T. Linnankivi report no disclosures relevant to the
manuscript. P. Isohanni has received research support from Foundation
for Pediatric Research. R. French and Y. Liu report no disclosures rele-
vant to the manuscript. M. Simonovic has received research support from
the US NIH grant GM097042. D. Söll received research support from
the US National Institute of General Medical Sciences (GM22854).
M. Somer, D. Muth-Pawlak, G. Corthals, A. Laari, E. Ylikallio,
M. Lähde, L. Valanne, T. Lönnqvist, H. Pihko, and A. Paetau report
no disclosures relevant to the manuscript. A. Lehesjoki has received
research support from the Folkhälsan Research Foundation. A. Suoma-
lainen has received research support from Sigrid Jusélius Foundation,
Academy of Finland, University of Helsinki, and Jane and Aatos Erkko
Foundation. H. Tyynismaa has received research support from Arvo and
Lea Ylppö Foundation, Orion Farmos Research Foundation, Sigrid
Jusélius Foundation, Academy of Finland, and University of Helsinki.
Go to Neurology.org for full disclosures.
Received November 17, 2014. Accepted in final form March 26, 2015.
REFERENCES
1. Koskinen T, Santavuori P, Sainio K, Lappi M, Kallio AK,
Pihko H. Infantile onset spinocerebellar ataxia with sen-
sory neuropathy: a new inherited disease. J Neurol Sci
1994;121:5056.
2. Nikali K, Suomalainen A, Saharinen J, et al. Infantile
onset spinocerebellar ataxia is caused by recessive muta-
tions in mitochondrial proteins Twinkle and Twinky.
Hum Mol Genet 2005;14:29812990.
3. Scheper GC, van der Klok T, van Andel RJ, et al. Mito-
chondrial aspartyl-tRNA synthetase deficiency causes leu-
koencephalopathy with brain stem and spinal cord
involvement and lactate elevation. Nat Genet 2007;39:
534539.
4. Elo JM, Yadavalli SS, Euro L, et al. Mitochondrial
phenylalanyl-tRNA synthetase mutations underlie fatal
infantile Alpers encephalopathy. Hum Mol Genet 2012;
21:45214529.
5. Ylikallio E, Suomalainen A. Mechanisms of mitochondrial
diseases. Ann Med 2012;44:4159.
Neurology 85 July 28, 2015 9
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
6. Edvardson S, Shaag A, Kolesnikova O, et al. Deleterious
mutation in the mitochondrial arginyl-transfer RNA syn-
thetase gene is associated with pontocerebellar hypoplasia.
Am J Hum Genet 2007;81:857862.
7. Somer M. Diagnostic criteria and genetics of the PEHO
syndrome. J Med Genet 1993;30:932936.
8. Sulonen AM, Ellonen P, Almusa H, et al. Comparison of
solution-based exome capture methods for next generation
sequencing. Genome Biol 2011;12:R94.
9. Emsley P, Lohkamp B, Scott WG, Cowtan K. Features
and development of coot. Acta Crystallogr D Biol Crys-
tallogr 2010;66:486501.
10. Palioura S, Sherrer RL, Steitz TA, Soll D, Simonovic M.
The human SepSecS-tRNASec complex reveals the mech-
anism of selenocysteine formation. Science 2009;325:
321325.
11. Yuan J, Palioura S, Salazar JC, et al. RNA-dependent con-
version of phosphoserine forms selenocysteine in eukar-
yotes and archaea. Proc Natl Acad Sci USA 2006;103:
1892318927.
12. Agamy O, Ben Zeev B, Lev D, et al. Mutations disrupting
selenocysteine formation cause progressive cerebello-
cerebral atrophy. Am J Hum Genet 2010;87:538544.
13. Bellinger FP, Raman AV, Reeves MA, Berry MJ. Regula-
tion and function of selenoproteins in human disease.
Biochem J 2009;422:1122.
14. Ben-Zeev B, Hoffman C, Lev D, et al. Progressive cere-
bellocerebral atrophy: a new syndrome with microcephaly,
mental retardation, and spastic quadriplegia. J Med Genet
2003;40:e96.
15. Harding BN. Progressive neuronal degeneration of child-
hood with liver disease (Alpers-Huttenlocher syndrome): a
personal review. J Child Neurol 1990;5:273287.
16. Hakonen AH, Isohanni P, Paetau A, Herva R,
Suomalainen A, Lonnqvist T. Recessive Twinkle muta-
tions in early onset encephalopathy with mtDNA deple-
tion. Brain 2007;130:30323040.
17. Naviaux RK, Nguyen KV. POLG mutations associated
with Alperssyndrome and mitochondrial DNA depletion.
Ann Neurol 2004;55:706712.
18. Salonen R, Somer M, Haltia M, Lorentz M, Norio R.
Progressive encephalopathy with edema, hypsarrhythmia,
and optic atrophy (PEHO syndrome). Clin Genet 1991;
39:287293.
19. Rankin J, Brown R, Dobyns WB, et al. Pontocerebellar
hypoplasia type 6: a British case with PEHO-like features.
Am J Med Genet A 2010;152A:20792084.
20. Budde BS, Namavar Y, Barth PG, et al. tRNA splicing
endonuclease mutations cause pontocerebellar hypoplasia.
Nat Genet 2008;40:11131118.
21. Kasher PR, Namavar Y, van Tijn P, et al. Impairment of
the tRNA-splicing endonuclease subunit 54 (tsen54) gene
causes neurological abnormalities and larval death in zebra-
fish models of pontocerebellar hypoplasia. Hum Mol
Genet 2011;20:15741584.
22. Serrano L, Fersht AR. Capping and alpha-helix stability.
Nature 1989;342:296299.
23. Schoenmakers E, Agostini M, Mitchell C, et al. Mutations
in the selenocysteine insertion sequence-binding protein 2
gene lead to a multisystem selenoprotein deficiency disor-
der in humans. J Clin Invest 2010;120:42204235.
24. Dumitrescu AM, Liao XH, Abdullah MS, et al. Mutations
in SECISBP2 result in abnormal thyroid hormone metab-
olism. Nat Genet 2005;37:12471252.
25. Bosl MR, Takaku K, Oshima M, Nishimura S,
Taketo MM. Early embryonic lethality caused by targeted
disruption of the mouse selenocysteine tRNA gene (trsp).
Proc Natl Acad Sci USA 1997;94:55315534.
26. Kasaikina MV, Hatfield DL, Gladyshev VN. Understand-
ing selenoprotein function and regulation through the use
of rodent models. Biochim Biophys Acta 2012;1823:
16331642.
27. Seiler A, Schneider M, Forster H, et al. Glutathione per-
oxidase 4 senses and translates oxidative stress into 12/15-
lipoxygenase dependent- and AIF-mediated cell death.
Cell Metab 2008;8:237248.
28. Soerensen J, Jakupoglu C, Beck H, et al. The role of thi-
oredoxin reductases in brain development. PLoS One
2008;3:e1813.
29. Wirth EK, Conrad M, Winterer J, et al. Neuronal seleno-
protein expression is required for interneuron development
and prevents seizures and neurodegeneration. FASEB J
2010;24:844852.
30. Wirth EK, Bharathi BS, Hatfield D, Conrad M,
Brielmeier M, Schweizer U. Cerebellar hypoplasia in mice
lacking selenoprotein biosynthesis in neurons. Biol Trace
Elem Res 2014;158:203210.
31. Schomburg L, Schweizer U, Holtmann B, Flohe L,
Sendtner M, Kohrle J. Gene disruption discloses role of
selenoprotein P in selenium delivery to target tissues. Bio-
chem J 2003;370:397402.
32. Hill KE, Zhou J, McMahan WJ, Motley AK, Burk RF.
Neurological dysfunction occurs in mice with targeted dele-
tion of the selenoprotein P gene. J Nutr 2004;134:157161.
10 Neurology 85 July 28, 2015
ª2015 American Academy of Neurology. Unauthorized reproduction of this article is prohibited.
DOI 10.1212/WNL.0000000000001787
published online June 26, 2015Neurology
Anna-Kaisa Anttonen, Taru Hilander, Tarja Linnankivi, et al.
lactate
Selenoprotein biosynthesis defect causes progressive encephalopathy with elevated
This information is current as of June 26, 2015
Services
Updated Information &
001787.full.html
http://www.neurology.org/content/early/2015/06/26/WNL.0000000000
including high resolution figures, can be found at:
Supplementary Material
0001787.DC1.html
http://www.neurology.org/content/suppl/2015/06/26/WNL.000000000
Supplementary material can be found at:
Subspecialty Collections
_genetics-mitochondrial_disorders
http://www.neurology.org//cgi/collection/mitochondrial_disorders_see
Mitochondrial disorders; see Genetics/Mitochondrial disorders
http://www.neurology.org//cgi/collection/infantile_spasms
Infantile spasms http://www.neurology.org//cgi/collection/all_pediatric
All Pediatric http://www.neurology.org//cgi/collection/all_genetics
All Genetics
following collection(s):
This article, along with others on similar topics, appears in the
Permissions & Licensing
http://www.neurology.org/misc/about.xhtml#permissions
its entirety can be found online at:
Information about reproducing this article in parts (figures,tables) or in
Reprints http://www.neurology.org/misc/addir.xhtml#reprintsus
Information about ordering reprints can be found online:
rights reserved. Print ISSN: 0028-3878. Online ISSN: 1526-632X.
1951, it is now a weekly with 48 issues per year. Copyright © 2015 American Academy of Neurology. All
® is the official journal of the American Academy of Neurology. Published continuously sinceNeurology
... Mild elevation of thyroidstimulating hormone (TSH) was observed in 2 of 4 EOPs. Anttonen et al. [8] conducted neuropathological analysis of 4 EOPs, and the results are shown in Table 3. 10 patients, including 6 EOPs and 4 LOPs, had detailed MRI description ( Table 4). The first MRI was normal in 2 EOPs and 3 LOPs. ...
... Some EOPs had epileptic seizures, and their EEG was mainly characterized by hypsarrhythmia and slow background with multifocal sharp waves. Sensory axonal neuropathy was verified by sural nerve biopsy and electroneuromyography in 3 EOPs [8]. Besides sensory axonal neuropathy, Pavlidou et al.'s case [10] also showed a mixed pattern containing both neurogenic and myopathic features in EMG. ...
... The neuropathological studies of other types are scarce or absent. Anttonen et al. [8] conducted neuropathological analysis of 4 patients with PCH2D caused by SEPSECS mutations and revealed that all 4 patients shared features of progressive neuronal degeneration and the white matter showed myelin loss and pallor with gliosis. All cases had a quite severe degeneration and atrophy of the brainstem and cerebellar cortex. ...
Article
Full-text available
Pontocerebellar hypoplasia type 2D (PCH2D) caused by SEPSECS gene mutations is very rare and only described in a few case reports. In this study, we analyzed the clinical features and imaging findings of these individuals, so as to provide references for the clinic. We reported a case of PCH2D caused by a new complex heterozygote mutation in SEPSECS gene, and reviewed the literatures to summarize the clinical features and imaging findings and compare the differences between early-onset patients (EOPs) and late-onset patients (LOPs). Of 23 PCH2D patients, 19 cases were early-onset and 4 cases were late-onset, with average ages of 4.1 ± 4.0 years and 21.8 ± 9.4 years, females were more prevalent (14/19). EOPs mainly distributed in Arab countries (10/14) and Finland (4/14), while LOPs in East Asia (3/3). EOPs develop severe initial symptoms at the average age of 4.1 ± 7.8 months or shortly after birth, while LOPs experienced mild developmental delay in infancy. Microcephaly (10/11), intellectual disability (10/11), decreased motor function (10/11), and spastic or dystonic quadriplegia (8/10) were the common clinical features of EOPs and LOPs. EOPs also presented with visual impairment (5/7), seizures (4/7), neonatal irritability/opisthotonus (3/7), tremors/myoclonus (3/7), dysmorphic features (3/7), and other symptoms. EOPs were characterized by cerebellar symptoms (4/4). Magnetic resonance imaging (MRI) revealed progressive cerebellar atrophy followed by less pronounced cerebral atrophy, and there was no pons atrophy in LOPs. Most patients of PCH2D were severe early-onset, and a few were late-onset with milder symptoms. EOPs and LOPs shared some common clinical features and MRI findings, but also had their own characteristics.
... Dyskinetic movement disorder characterizes the subgroup type 2. PCH2D may span axonal neuropathy, myopathy, epilepsy or epileptic encephalopathy featuring West syndrome, optic nerve hypoplasia, and high blood lactate. [3][4][5] In addition, milder neurodegenerative phenotype characterized by earlyonset ataxia with microcephaly and progressive cerebellar atrophy has been described in 3 patients. 6,7 Our patient had a unique combination of pyramidalextrapyramidal syndrome and optic nerve hypoplasia with TCC without basal ganglia, cerebellum, and midbrain anomalies. ...
... An analysis of the few published pathogenic SEPSECS variants indicated that different types of mutations are spread along the gene, thus not clarifying the genotype-phenotype correlations. [3][4][5][6][7] Reasons for phenotypic variability remain to be explained. ...
Article
Full-text available
Objectives To report a novel association between pathogenic variants in the SEPSECS gene and complex movement disorder with thin corpus callosum (TCC). Methods Clinical exome sequencing was performed in an adult patient with a genetically unsolved neurodegenerative disorder. The main clinical, neuroimaging, and genetic data were described. Results The c.865C > T (p.P289S) and c.1297T > C (p.Y433H) missense variants in SEPSECS (NM_016,955.3) were discovered. Discussion This case represents a novel form of early-onset pyramidal syndrome with optic nerve hypoplasia, which slowly evolved to extrapyramidal syndrome featuring dystonia-parkinsonism, associated with TCC, caused by SEPSECS pathogenic variants. This form enlarges the group of the so-called pyramidal-extrapyramidal syndromes, as well as complex hereditary spastic paraparesis with TCC.
... Since mice with neuronal selenoproteins deficiency show cerebellar hypoplasia, it seems selenoproteins play a crucial role in brain development [33]. Selenoproteins are also involved in antioxidant defense, and reduced selenoproteins levels could damage organs with high mitochondrial activity since mitochondria are one the primary sources of oxidative stress in cells [34]. ...
Article
Full-text available
Background Pontocerebellar hypoplasia is an umbrella term describing a heterogeneous group of prenatal neurodegenerative disorders mostly affecting the pons and cerebellum, with 17 types associated with 25 genes. However, some types of PCH lack sufficient information, which highlights the importance of investigating and introducing more cases to further elucidate the clinical, radiological, and biochemical features of these disorders. The aim of this study is to provide an in-depth review of PCH and to identify disease genes and their inheritance patterns in 12 distinct Iranian families with clinically confirmed PCH. Methods Cases included in this study were selected based on their phenotypic and genetic information available at the Center for Comprehensive Genetic Services. Whole-exome sequencing (WES) was used to discover the underlying genetic etiology of participants' problems, and Sanger sequencing was utilized to confirm any suspected alterations. We also conducted a comprehensive molecular literature review to outline the genetic features of the various subtypes of PCH. Results This study classified and described the underlying etiology of PCH into three categories based on the genes involved. Twelve patients also were included, eleven of whom were from consanguineous parents. Ten different variations in 8 genes were found, all of which related to different types of PCH. Six novel variations were reported, including SEPSECS, TSEN2, TSEN54, AMPD2, TOE1, and CLP1. Almost all patients presented with developmental delay, hypotonia, seizure, and microcephaly being common features. Strabismus and elevation in lactate levels in MR spectroscopy were novel phenotypes for the first time in PCH types 7 and 9. Conclusions This study merges previously documented phenotypes and genotypes with unique novel ones. Due to the diversity in PCH, we provided guidance for detecting and diagnosing these heterogeneous groups of disorders. Moreover, since certain critical conditions, such as spinal muscular atrophy, can be a differential diagnosis, providing cases with novel variations and clinical findings could further expand the genetic and clinical spectrum of these diseases and help in better diagnosis. Therefore, six novel genetic variants and novel clinical and paraclinical findings have been reported for the first time. Further studies are needed to elucidate the underlying mechanisms and potential therapeutic targets for PCH.
... Selenoproteins are potent antioxidants and play a key role in controlling cellular ROS levels. Notably, mutations in selenoprotein biosynthesis machinery are associated with neurodevelopmental disorders (53)(54)(55)(56)(57). More recently, mutations in ALKBH8 have been identified as the underlying cause of autosomal-recessive intellectual disability in multiple families (58)(59)(60)(61). ...
Preprint
Full-text available
Post-transcriptional modification of RNA regulates gene expression at multiple levels. ALKBH8 is a tRNA modifying enzyme that methylates wobble uridines in specific tRNAs to modulate translation. Through methylation of tRNA-selenocysteine, ALKBH8 promotes selenoprotein synthesis and regulates redox homeostasis. Pathogenic variants in ALKBH8 have been linked to intellectual disability disorders in the human population, but the role of ALKBH8 in the nervous system is unknown. Through in vivo studies in Drosophila , we show that ALKBH8 controls oxidative stress in the brain to restrain synaptic growth and support learning and memory. ALKBH8 null animals lack wobble uridine methylation and exhibit a global reduction in protein synthesis, including a specific decrease in selenoprotein levels. Loss of ALKBH8 or independent disruption of selenoprotein synthesis results in ectopic synapse formation. Genetic expression of antioxidant enzymes fully suppresses synaptic overgrowth in ALKBH8 null animals, confirming oxidative stress as the underlying cause of dysregulation. ALKBH8 animals also exhibit associative learning and memory impairments that are reversed by pharmacological antioxidant treatment. Together, these findings demonstrate the critical role of tRNA modification in redox homeostasis in the nervous system and reveal antioxidants as a potential therapy for ALKBH8-associated intellectual disability. Significance Statement tRNA modifying enzymes are emerging as important regulators of nervous system development and function due to their growing links to neurological disorders. Yet, their roles in the nervous system remain largely elusive. Through in vivo studies in Drosophila , we link tRNA methyltransferase-regulated selenoprotein synthesis to synapse development and associative memory. These findings demonstrate the key role of tRNA modifiers in redox homeostasis during nervous system development and highlight the potential therapeutic benefit of antioxidant-based therapies for cognitive disorders linked to dysregulation of tRNA modification.
... Data from genome-wide association studies suggested that common variants in the ZNF595 gene may be associated with SCZ and SCZ-related traits [30]. Pathogenic mutations in selenocysteine synthase (SEPSECS) cause neurodevelopmental disorders [31][32][33]. While using sequencing data analysis, Laan, L identified regional epigenetic changes in the transcription factor gene ZNF700, which is relevant in Down syndrome brain development, providing a novel framework for further studies on epigenetic changes and transcriptional dysregulation during chromosome 21 neurogenesis [34]. ...
Article
Full-text available
Background: As the most common arrhythmia, atrial fibrillation (AF) is associated with a significantly increased risk of stroke, which causes high disability and mortality. To date, the underlying mechanism of stroke occurring after AF remains unclear. Herein, we studied hub genes and regulatory pathways involved in AF and secondary stroke and aimed to reveal biomarkers and therapeutic targets of AF-related stroke. Methods: The GSE79768 and GSE58294 datasets were used to analyze AF- and stroke-related differentially expressed genes (DEGs) to obtain a DEG1 dataset. Weighted correlation network analysis (WGCNA) was used to identify modules associated with AF-related stroke in GSE66724 (DEG2). DEG1 and DEG2 were merged, and hub genes were identified based on protein-protein interaction networks. Gene Ontology terms were used to analyze the enriched pathways. The GSE129409 and GSE70887 were applied to construct a circRNA-miRNA-mRNA network in AF-related stroke. Hub genes were verified in patients using quantitative real-time polymerase chain reaction (qRT-PCR). Results: We identified 3,132 DEGs in blood samples and 253 DEGs in left atrial specimens. Co-expressed hub genes of EIF4E3, ZNF595, ZNF700, MATR3, ACKR4, ANXA3, SEPSECS-AS1, and RNF166 were significantly associated with AF-related stroke. The hsa_circ_0018657/hsa-miR-198/EIF4E3 pathway was explored as the regulating axis in AF-related stroke. The qRT-PCR results were consistent with the bioinformatic analysis. Conclusions: Hub genes EIF4E3, ZNF595, ZNF700, MATR3, ACKR4, ANXA3, SEPSECS-AS1, and RNF166 have potential as novel biomarkers and therapeutic targets in AF-related stroke. The hsa_circ_0018657/hsa-miR-198/EIF4E3 axis could play an important role regulating the development of AF-related stroke.
Article
Significance: Physiological levels of reactive oxygen and nitrogen species (ROS/RNS) function as fundamental messengers for many cellular and developmental processes in the cardiovascular system. ROS/RNS involved in cardiac redox-signaling originate from diverse sources, and their levels are tightly controlled by key endogenous antioxidant systems that counteract their accumulation. However, dysregulated redox-stress resulting from inefficient removal of ROS/RNS leads to inflammation, mitochondrial dysfunction, and cell death, contributing to the development and progression of cardiovascular disease (CVD). Recent advances: Basic and clinical studies demonstrate the critical role of selenium (Se) and selenoproteins (unique proteins that incorporate Se into their active site in the form of the 21st proteinogenic amino acid selenocysteine, Sec), including glutathione peroxidase (GPX) and thioredoxin reductase (TXNRD), in cardiovascular redox homeostasis, representing a first-line enzymatic antioxidant defence of the heart. Increasing attention has been paid to emerging selenoproteins in the endoplasmic reticulum (ER) (i.e., a multifunctional intracellular organelle whose disruption triggers cardiac inflammation and oxidative stress, leading to multiple CVD), which are crucially involved in redox balance, antioxidant activity, calcium (Ca2+) and ER homeostasis. Critical issues: This review focuses on endogenous antioxidant strategies with therapeutic potential, particularly selenoproteins, which are very promising but deserve more detailed and clinical studies. Future directions: The importance of selective selenoproteins in embryonic development and the consequences of their mutations and inborn errors highlight the need to improve knowledge of their biological function in myocardial redox signaling. This could facilitate the development of personalized approaches for the diagnosis, prevention, and treatment of CVD.
Article
Full-text available
Selenoproteins are a group of proteins containing selenium in the form of selenocysteine (Sec, U) as the 21st amino acid coded in the genetic code. Their synthesis depends on dietary selenium uptake and a common set of cofactors. Selenoproteins accomplish diverse roles in the body and cell processes by acting, for example, as antioxidants, modulators of the immune function, and detoxification agents for heavy metals, other xenobiotics, and key compounds in thyroid hormone metabolism. Although the functions of all this protein family are still unknown, several disorders in their structure, activity, or expression have been described by researchers. They concluded that selenium or cofactors deficiency, on the one hand, or the polymorphism in selenoproteins genes and synthesis, on the other hand, are involved in a large variety of pathological conditions, including type 2 diabetes, cardiovascular, muscular, oncological, hepatic, endocrine, immuno-inflammatory, and neurodegenerative diseases. This review focuses on the specific roles of selenoproteins named after letters of the alphabet in medicine, which are less known than the rest, regarding their implications in the pathological processes of several prevalent diseases and disease prevention.
Article
Progressive encephalopathy with edema, hypsarrhythmia, and optic atrophy (PEHO) and PEHO-like syndromes are very rare infantile disorders characterized by profound intellectual disability, hypotonia, convulsions, optic, and progressive brain atrophy. Many causative genes for PEHO and PEHO-like syndromes have been identified including CCDC88A. So far, only five patients from two unrelated families with biallelic CCDC88A variants have been reported in the literature. Herein, we describe a new family from Egypt with a lethal epileptic encephalopathy. Our patient was the youngest child born to a highly consanguineous couple and had a family history of five deceased sibs with the same condition. She presented with postnatal microcephaly, poor visual responsiveness, and epilepsy. Her brain MRI showed abnormal cortical gyration with failure of opercularization of the insula, hypogenesis of corpus callosum, colpocephaly, reduced white matter, hypoplastic vermis, and brain stem. Whole exome sequencing identified a new homozygous frameshift variant in CCDC88A gene (c.1795_1798delACAA, p.Thr599ValfsTer4). Our study presents the third reported family with this extremely rare disorder. We also reviewed all described cases to better refine the phenotypic spectrum associated with biallelic loss of function variants in the CCDC88A gene.
Article
Expression of selenoproteins is widespread in neurons of the central nervous system. There is continuous evidence presented over decades that low levels of selenium or selenoproteins are linked to seizures and epilepsy indicating a failure of the inhibitory system. Many developmental processes in the brain depend on the thyroid hormone T3. T3 levels can be locally increased by the action of iodothyronine deiodinases on the prohormone T4. Since deiodinases are selenoproteins, it is expected that selenoprotein deficiency may affect development of the central nervous system. Studies in genetically modified mice or clinical observations of patients with rare diseases point to a role of selenoproteins in brain development and degeneration. In particular selenoprotein P is central to brain function by virtue of its selenium transport function into and within the brain. We summarize which selenoproteins are essential for the brain, which processes depend on selenoproteins, and what is known about genetic deficiencies of selenoproteins in humans. This review is not intended to cover the potential influence of selenium or selenoproteins on major neurodegenerative disorders in human.
Thesis
Mutations in the human mitochondrial genome are often associated with severe neuromuscular disorders. The first part of my thesis project consisted in the development of a therapeutic strategy based on the mitochondrial import of RNA molecules. I demonstrated that the stable expression of recombinant RNA molecules in human cells induced the decrease of the pathogenic mutation load in mitochondrial DNA. In the second part, I developed a nex method, CoLoC-seq, for the large-scale identification of RNA species localized in the mitochondria. By applying this method to human cells, I confirmed the mitochondrial targeting of some non-coding cytosolic RNAs and identified new potentially imported RNAs. These data will broaden the knowledge on the pathway of RNA targeting into the mitochondria, its mechanisms and regulation, and will allow optimization of the therapeutic strategies based on RNA import.
Article
Full-text available
Making Selenocysteine In humans, selenocysteine is the only amino acid that lacks its own transfer RNA (tRNA) synthetase and is synthesized on its cognate tRNA. The process involves mischarging of tRNA sec with serine, phosphorylation of the serine, and then conversion of the phosphoserine into selenocysteine by the enzyme SepSecS using selenophosphate as the selenium donor. Palioura et al. (p. 321 ) now provide insight into the mechanism of selenocysteine formation, based on the crystal structure of human tRNA sec complexed with SepSecS, phosphoserine, and thiophosphate, together with in vivo and in vitro activity assays. Binding of tRNA sec to SepSecS is required to properly orient phosphoserine attached to tRNA sec for pyroxidal phosphate–based catalysis.
Article
Full-text available
Selenium exerts many, if not most, of its physiological functions as a selenocysteine moiety in proteins. Selenoproteins are involved in many biochemical processes including regulation of cellular redox state, calcium homeostasis, protein biosynthesis, and degradation. A neurodevelopmental syndrome called progressive cerebello-cortical atrophy (PCCA) is caused by mutations in the selenocysteine synthase gene, SEPSECS, demonstrating that selenoproteins are essential for human brain development. While we have shown that selenoproteins are required for correct hippocampal and cortical interneuron development, little is known about the functions of selenoproteins in the cerebellum. Therefore, we have abrogated neuronal selenoprotein biosynthesis by conditional deletion of the gene encoding selenocysteyl tRNA([Ser]Sec) (gene symbol Trsp). Enzymatic activity of cellular glutathione peroxidase and cytosolic thioredoxin reductase is reduced in cerebellar extracts from Trsp-mutant mice. These mice grow slowly and fail to gain postural control or to coordinate their movements. Histological analysis reveals marked cerebellar hypoplasia, associated with Purkinje cell death and decreased granule cell proliferation. Purkinje cell death occurs along parasagittal stripes as observed in other models of Purkinje cell loss. Neuron-specific inactivation of glutathione peroxidase 4 (Gpx4) used the same Cre driver phenocopies tRNA([Ser]Sec) mutants in several aspects: cerebellar hypoplasia, stripe-like Purkinje cell loss, and reduced granule cell proliferation. Parvalbumin-expressing GABAergic interneurons (stellate and/or basket cells) are virtually absent in tRNA([Ser]Sec)-mutant mice, while some remained in Gpx4-mutant mice. Our data show that selenoproteins are specifically required in postmitotic neurons of the developing cerebellum, thus providing a rational explanation for cerebellar hypoplasia as occurring in PCCA patients.
Article
Full-text available
Next-generation sequencing has turned out to be a powerful tool to uncover genetic basis of childhood mitochondrial disorders. We utilized whole-exome analysis and discovered novel compound heterozygous mutations in FARS2 (mitochondrial phenylalanyl transfer RNA synthetase), encoding the mitochondrial phenylalanyl transfer RNA (tRNA) synthetase (mtPheRS) in two patients with fatal epileptic mitochondrial encephalopathy. The mutations affected highly conserved amino acids, p.I329T and p.D391V. Recently, a homozygous FARS2 variant p.Y144C was reported in a Saudi girl with mitochondrial encephalopathy, but the pathogenic role of the variant remained open. Clinical features, including postnatal onset, catastrophic epilepsy, lactic acidemia, early lethality and neuroimaging findings of the patients with FARS2 variants, resembled each other closely, and neuropathology was consistent with Alpers syndrome. Our structural analysis of mtPheRS predicted that p.I329T weakened ATP binding in the aminoacylation domain, and in vitro studies with recombinant mutant protein showed decreased affinity of this variant to ATP. Furthermore, p.D391V and p.Y144C were predicted to disrupt synthetase function by interrupting the rotation of the tRNA anticodon stem-binding domain from a closed to an open form. In vitro characterization indicated reduced affinity of p.D391V mutant protein to phenylalanine, whereas p.Y144C disrupted tRNA binding. The stability of p.I329T and p.D391V mutants in a refolding assay was impaired. Our results imply that the three FARS2 mutations directly impair aminoacylation function and stability of mtPheRS, leading to a decrease in overall tRNA charging capacity. This study establishes a new genetic cause of infantile mitochondrial Alpers encephalopathy and reports a new mitochondrial aminoacyl-tRNA synthetase as a cause of mitochondrial disease.
Article
Full-text available
Techniques enabling targeted re-sequencing of the protein coding sequences of the human genome on next generation sequencing instruments are of great interest. We conducted a systematic comparison of the solution-based exome capture kits provided by Agilent and Roche NimbleGen. A control DNA sample was captured with all four capture methods and prepared for Illumina GAII sequencing. Sequence data from additional samples prepared with the same protocols were also used in the comparison. We developed a bioinformatics pipeline for quality control, short read alignment, variant identification and annotation of the sequence data. In our analysis, a larger percentage of the high quality reads from the NimbleGen captures than from the Agilent captures aligned to the capture target regions. High GC content of the target sequence was associated with poor capture success in all exome enrichment methods. Comparison of mean allele balances for heterozygous variants indicated a tendency to have more reference bases than variant bases in the heterozygous variant positions within the target regions in all methods. There was virtually no difference in the genotype concordance compared to genotypes derived from SNP arrays. A minimum of 11× coverage was required to make a heterozygote genotype call with 99% accuracy when compared to common SNPs on genome-wide association arrays. Libraries captured with NimbleGen kits aligned more accurately to the target regions. The updated NimbleGen kit most efficiently covered the exome with a minimum coverage of 20×, yet none of the kits captured all the Consensus Coding Sequence annotated exons.
Article
Full-text available
Mitochondria are essential organelles with multiple functions, the most well known being the production of adenosine triphosphate (ATP) through oxidative phosphorylation (OXPHOS). The mitochondrial diseases are defined by impairment of OXPHOS. They are a diverse group of diseases that can present in virtually any tissue in either adults or children. Here we review the main molecular mechanisms of mitochondrial diseases, as presently known. A number of disease-causing genetic defects, either in the nuclear genome or in the mitochondria's own genome, mitochondrial DNA (mtDNA), have been identified. The most classical genetic defect causing mitochondrial disease is a mutation in a gene encoding a structural OXPHOS subunit. However, mitochondrial diseases can also arise through impaired mtDNA maintenance, defects in mitochondrial translation factors, and various more indirect mechanisms. The putative consequences of mitochondrial dysfunction on a cellular level are discussed.
Article
Full-text available
Pontocerebellar hypoplasia (PCH) represents a group (PCH1-6) of neurodegenerative autosomal recessive disorders characterized by hypoplasia and/or atrophy of the cerebellum, hypoplasia of the ventral pons, progressive microcephaly and variable neocortical atrophy. The majority of PCH2 and PCH4 cases are caused by mutations in the TSEN54 gene; one of the four subunits comprising the tRNA-splicing endonuclease (TSEN) complex. We hypothesized that TSEN54 mutations act through a loss of function mechanism. At 8 weeks of gestation, human TSEN54 is expressed ubiquitously in the brain, yet strong expression is seen within the telencephalon and metencephalon. Comparable expression patterns for tsen54 are observed in zebrafish embryos. Morpholino (MO) knockdown of tsen54 in zebrafish embryos results in loss of structural definition in the brain. This phenotype was partially rescued by co-injecting the MO with human TSEN54 mRNA. A developmental patterning defect was not associated with tsen54 knockdown; however, an increase in cell death within the brain was observed, thus bearing resemblance to PCH pathophysiology. Additionally, N-methyl-N-nitrosourea mutant zebrafish homozygous for a tsen54 premature stop-codon mutation die within 9 days post-fertilization. To determine whether a common disease pathway exists between TSEN54 and other PCH-related genes, we also monitored the effects of mitochondrial arginyl-tRNA synthetase (rars2; PCH1 and PCH6) knockdown in zebrafish. Comparable brain phenotypes were observed following the inhibition of both genes. These data strongly support the hypothesis that TSEN54 mutations cause PCH through a loss of function mechanism. Also we suggest that a common disease pathway may exist between TSEN54- and RARS2-related PCH, which may involve a tRNA processing-related mechanism.
Article
Full-text available
Selenium, a trace element that is fundamental to human health, is incorporated into some proteins as selenocysteine (Sec), generating a family of selenoproteins. Sec incorporation is mediated by a multiprotein complex that includes Sec insertion sequence-binding protein 2 (SECISBP2; also known as SBP2). Here, we describe subjects with compound heterozygous defects in the SECISBP2 gene. These individuals have reduced synthesis of most of the 25 known human selenoproteins, resulting in a complex phenotype. Azoospermia, with failure of the latter stages of spermatogenesis, was associated with a lack of testis-enriched selenoproteins. An axial muscular dystrophy was also present, with features similar to myopathies caused by mutations in selenoprotein N (SEPN1). Cutaneous deficiencies of antioxidant selenoenzymes, increased cellular ROS, and susceptibility to ultraviolet radiation-induced oxidative damage may mediate the observed photosensitivity. Reduced levels of selenoproteins in peripheral blood cells were associated with impaired T lymphocyte proliferation, abnormal mononuclear cell cytokine secretion, and telomere shortening. Paradoxically, raised ROS in affected subjects was associated with enhanced systemic and cellular insulin sensitivity, similar to findings in mice lacking the antioxidant selenoenzyme glutathione peroxidase 1 (GPx1). Thus, mutation of SECISBP2 is associated with a multisystem disorder with defective biosynthesis of many selenoproteins, highlighting their role in diverse biological processes.
Article
Alpers' syndrome is a fatal neurogenetic disorder first described more than 70 years ago. It is an autosomal recessive, developmental mitochondrial DNA depletion disorder characterized by deficiency in mitochondrial DNA polymerase gamma (POLG) catalytic activity, refractory seizures, neurodegeneration, and liver disease. In two unrelated pedigrees of Alpers' syndrome, each affected child was found to carry a homozygous mutation in exon 17 of the POLG locus that led to a Glu873Stop mutation just upstream of the polymerase domain of the protein. In addition, each affected child was heterozygous for the G1681A mutation in exon 7 that led to an Ala467Thr substitution in POLG, within the linker region of the protein.
Article
Selenium (Se) is an essential micronutrient. Its biological functions are associated with selenoproteins, which contain this trace element in the form of the 21st amino acid, selenocysteine. Genetic defects in selenocysteine insertion into proteins are associated with severe health issues. The consequences of selenoprotein deficiency are more variable, with several selenoproteins being essential, and several showing no clear phenotypes. Much of these functional studies benefited from the use of rodent models and diets employing variable levels of Se. This review summarizes the data obtained with these models, focusing on mouse models with targeted expression of individual selenoproteins and removal of individual, subsets or all selenoproteins in a systemic or organ-specific manner. This article is part of a Special Issue entitled: Cell Biology of Metals.