ArticlePDF Available

Mild form of Zellweger Spectrum Disorders (ZSD) due to variants in PEX1: Detailed clinical investigation in a 9-years-old female

Authors:

Abstract and Figures

Peroxisomal biogenesis disorders (PBD) are rare autosomal recessive disorders with various degrees of severity caused by hypomorphic mutations in 13 different peroxin (PEX) genes. In this study, we report the clinical and molecular characterization of a 9-years-old female presenting an apparently isolated pre-lingual sensorineural hearing loss (SNHL) and early onset Retinitis Pigmentosa (RP) that may clinically overlap with Usher syndrome. Genetic testing by clinical exome sequencing identified two variants in PEX1: the missense variant c.274G > C; p.(Val92Leu) that was already reported in a PBD patient, and the variant c.2140_2145dup; p.(Ser714_Gln715dup) which is a novel, non-frameshift variant, absent in control databases. On the basis of the molecular analysis, a thorough clinical examination revealed nail and dental abnormalities, a mild cognitive impairment, learning disabilities and poor feeding, apart from the retinal and audiological features initially identified. The clinical and molecular findings led us to the diagnosis of a mild form of PBD. This study further emphasizes that mild forms of PBD can be a differential diagnosis of Usher syndrome and suggests that patients with mild cognitive impairment associated to visual and hearing loss should perform a comprehensive mutation screening that includes PEX genes.
Content may be subject to copyright.
Contents lists available at ScienceDirect
Molecular Genetics and Metabolism Reports
journal homepage: www.elsevier.com/locate/ymgmr
Case Report
Mild form of Zellweger Spectrum Disorders (ZSD) due to variants in PEX1:
Detailed clinical investigation in a 9-years-old female
Maria Rosaria Barillari
a,1,
, Marianthi Karali
b,c,1
, Valentina Di Iorio
d
, Maria Contaldo
e
,
Vincenzo Piccolo
f
, Maria Esposito
g
, Giuseppe Costa
a
, Giuseppe Argenziano
f
, Rosario Serpico
e
,
Marco Carotenuto
g
, Gerarda Cappuccio
b,h
, Sandro Ban
b,c
, Paolo Melillo
d
, Francesca Simonelli
d
a
Division of Phoniatrics and Audiology, Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio
4, 80138 Naples, Italy
b
Telethon Institute of Genetics and Medicine, Pozzuoli, Via Campi Flegrei 34, 80078 Pozzuoli, Italy
c
Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
d
Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania Luigi Vanvitelli, Via Pansini 5, 80131 Naples, Italy
e
Dental Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania Luigi Vanvitelli, Via L. De Crecchio 6, 80138 Naples, Italy
f
Pediatric Dermatology, Dermatology Unit, University of Campania Luigi Vanvitelli, Via Pansini 5, 80131 Naples, Italy
g
Clinic of Child and Adolescent Neuropsychiatry, Department of Mental Health, Physical and Preventive Medicine, University of Campania "Luigi Vanvitelli", Via Pansini 5,
80131 Naples, Italy
h
Department of Translational Medicine, Section of Paediatrics, Federico II University, Via Pansini 5, 80131 Naples, Italy
ARTICLE INFO
Keywords:
Sensorineural hearing loss
Retinitis pigmentosa
Enamel defects
PEX genes
Peroxisomal biogenesis disorders
Mild Zellweger syndrome
ABSTRACT
Peroxisomal biogenesis disorders (PBD) are rare autosomal recessive disorders with various degrees of severity
caused by hypomorphic mutations in 13 dierent peroxin (PEX) genes. In this study, we report the clinical and
molecular characterization of a 9-years-old female presenting an apparently isolated pre-lingual sensorineural
hearing loss (SNHL) and early onset Retinitis Pigmentosa (RP) that may clinically overlap with Usher syndrome.
Genetic testing by clinical exome sequencing identied two variants in PEX1: the missense variant c.274G > C;
p.(Val92Leu) that was already reported in a PBD patient, and the variant c.2140_2145dup; p.
(Ser714_Gln715dup) which is a novel, non-frameshift variant, absent in control databases. On the basis of the
molecular analysis, a thorough clinical examination revealed nail and dental abnormalities, a mild cognitive
impairment, learning disabilities and poor feeding, apart from the retinal and audiological features initially
identied. The clinical and molecular ndings led us to the diagnosis of a mild form of PBD. This study further
emphasizes that mild forms of PBD can be a dierential diagnosis of Usher syndrome and suggests that patients
with mild cognitive impairment associated to visual and hearing loss should perform a comprehensive mutation
screening that includes PEX genes.
1. Introduction
Peroxisomal biogenesis disorders (PBDs) are a group of autosomal
recessive disorders caused by mutations in one of the thirteen Peroxin
(PEX) genes. PBDs are caused by partial or generalized defects in per-
oxisome biogenesis [1]. These organelles are present in almost all eu-
karyotic cells and play an indispensable role in many biochemical
pathways including the metabolism of branched chain and very long
chain fatty acids, ether lipids, polyamines, amino acids and glyoxylate
[2,3]. Peroxisomal dysfunction leads to multisystem disease that in-
cludes neurological, visual and hearing symptoms [4].
PBDs are divided into two main types: the Rhizomelic
Chondrodysplasia Punctata type 1 and the Zellweger spectrum disorder
(PBD-ZSD) which ranges from severe (Zellweger Syndrome, ZS) to
https://doi.org/10.1016/j.ymgmr.2020.100615
Received 21 March 2020; Received in revised form 5 June 2020; Accepted 6 June 2020
Abbreviations: PBD, Peroxisomal biogenesis disorders; SNHL, sensorineural hearing loss; RP, retinitis pigmentosa; PEX, peroxin; ZSD, Zellweger spectrum disorder;
ZS, Zellweger Syndrome; HS, Heimler syndrome; BCVA, Best Corrected Visual Acuity; GVF, Goldmann Visual Field; ERG, full-eld electroretinogram; OCT, optical
coherence tomography; FAF, color fundus and fundus autouorescence; TEOAE, Transient-Evoked Otoacustic Emission; ABR, Auditory Brainstem Responses; PTA,
Pure Tone Average; WISC-IV, Wechsler Intelligence Scale for Children (4th Edition); CDI, Childrens Depression Inventory; VLCFA, Very Long Chain Fatty Acid
Corresponding author.
E-mail address: mariarosaria.barillari@unicampania.it (M.R. Barillari).
1
These authors contributed equally to this work.
Molecular Genetics and Metabolism Reports 24 (2020) 100615
2214-4269/ © 2020 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/BY/4.0/).
T
intermediate (Neonatal Adreno-LeukoDystrophy) and mild (Infantile
Refsum Disease) phenotypes [5]. The PBD-ZSD is a clinically hetero-
geneous group with a continuum of disease severity. In particular, the
clinical presentation of the most severe form, namely ZS (OMIM:
614872), is characterized by craniofacial abnormalities, neuronal mi-
gration defects, leukodystrophy, absence of language, cognitive and
psychomotor delay, renal and liver diseases, hypotonia, hearing loss
and vision problems including cataracts and/or retinal abnormalities
[6]. Children with this condition do not develop properly and usually
die before one year of age [7], while patients with intermediate and
mild phenotypes can live into adulthood because their clinical features
(e.g. sensorineural hearing loss, retinal diseases, leukodystrophy and
cognitive delay) are less pronounced compared to those of ZS. Patho-
genic variants in PEX1 are the most common cause of PBD-ZSD and
have been associated with various degrees of disease severity [811].
On the other hand, the Heimler Syndrome (HS) is considered the
mildest end of PBD-ZSD spectrum of disorders [12]. HS is caused by
hypomorphic mutations in PEX1 and PEX6 genes and is characterized
by severe to profound bilateral sensorineural deafness, enamel defects
and nail abnormalities such as Beau's lines and punctate leukonychia
[1215]. Macular dystrophy has also been reported in the context of the
HS
16
.
The clinical heterogeneity of PBD-ZSD renders patients' diagnosis
challenging, especially in the case of very mild, late-onset forms that
may overlap with other syndromic phenotypes, in particular Usher
syndrome. This may delay diagnosis and proper management of the
disease.
Here, we report the clinical and molecular characterization of a 9-
years-old female presenting an apparently isolated sensorineural
hearing loss (SNHL) and early onset atypical Retinitis Pigmentosa (RP).
Clinical exome sequencing identied two biallelic variants in the PEX1
gene. A thorough clinical reevaluation led to the diagnosis of a mild
form of PBD-ZSD. We therefore suggest that patients with mild cogni-
tive impairment associated to visual and hearing loss should perform a
comprehensive mutation screening that includes PEX genes.
2. Materials and methods
Written informed consent for research and publication was obtained
by the family prior to participation of the subject to the current study.
All the procedures of this study were in complete accordance with the
Declaration of Helsinki on ethical principles for medical research in-
volving human subjects (2014).
The patient underwent a multidisciplinary clinical examination. It
included ophthalmological and audiological tests as well as pediatric,
dental, dermatological and neuropsychological evaluation and standard
biochemical tests.
2.1. Ophthalmological evaluation
Best Corrected Visual Acuity (BCVA), slit lamp anterior segment
examination, Goldmann Visual Field (GVF), fundus examination, full-
eld electroretinogram (ERG), optical coherence tomography (OCT),
color fundus and fundus autouorescence (FAF) were performed.
2.2. Audiological evaluation
The audiological assessment was obtained by the use of Otoacustic
Emission (OAE), in terms of Transient-Evoked Otoacustic Emission
(TEOAE), Auditory Brainstem Responses (ABR), liminar pure tone
audiometry with evaluation of the Pure Tone Average (PTA) and
Impedance test composed by tympanometry and stapedial reexes.
2.3. Neuropsychological evaluation
This clinical examination was performed by a trained child
psychiatrist and included an interview with the patient, clinical ob-
servation of her behavior and the administration of validated tests and
questionnaires such as SCARED and WISC-IV.
2.4. Dental evaluation
An extra- and intra-oral examination was performed to evaluate the
oral mucosal integrity and teeth status. An X-ray orthopantomography
was combined with the clinical evaluation to identify any dental
agenesis, dental malocclusions and other abnormalities in the shape
and/or structure of the teeth.
2.5. Dermatological examination
A total body examination was performed including inspection of
nails and hair. Further details about the above-mentioned tests are
provided in Supplementary Materials.
2.6. Biochemical analysis
Measurement of plasmatic Very Long Chain Fatty Acid (VLCFA) and
branched-chain fatty acid was performed by a specialized laboratory
using capillary gas chromatography/mass spectrometry of penta-
uorobenzyl bromide fatty acid esters as described elsewhere [17,18].
2.7. Clinical exome sequencing and segregation analysis
The proband's genomic DNA sample underwent a panel-based Next
Generation Sequencing using the ClearSeq Inherited Disease Panel
(Agilent) that allows the screening of more than 2700 genes known to
cause human inherited disorders. Sequencing data were analysed as
previously described [19]. Selected variations were validated by Sanger
sequencing in the patient and parents to assess proper segregation.
Further details are provided in Supplementary Materials.
3. Results
The patient was born at 40 weeks of gestation to unrelated parents
with no prior pregnancies. The Apgar index at birth was 8/10. No
dysmorphic features were evident, and no low muscle tone was re-
ported.
At 3 months of age she presented a bilateral sensorineural hearing
loss, diagnosed by the use of TEOAE and ABR. The auditory decit was
treated by digital hearing aids and speech therapy. Annual audiological
follow-ups were regularly performed.
The patient was clinically evaluated by our multidisciplinary team
for the rst time at the age of 9 because of a suspicion of an inherited
retinal disease associated to SNHL.
3.1. General pediatric evaluation
In the last pediatric evaluation performed at 9 years of age the
subject had a height of 128.6 cm (2.17 SD), an OFC of 49.3 cm (3.7
SD) and weighed 22.9 Kg (2.6 SD). Her low weight was associated
also with a signicant food selectivity and poor feeding for which a
specic behavioral feeding program was indicated.
The values of the routine hematochemical analysis were within the
normal range. Liver function tests showed normal levels of glutamic
oxaloacetic transaminase (GOT) and serum glutamic-pyruvic transa-
minase (GPT) at 9 years, respectively of 21.58 U/L (N·V: < 42 U/L) and
21.23 U/L (N·V: < 41). Similarly, alkaline phosphatase, albumin,
creatinine and total protein values were normal. Total bile acids were
normal at the age of 9 while total bilirubin resulted weakly elevated at
the last check (1.49 mg%; N·V: < 1).
Abdominal ultrasounds revealed no major renal or liver abnormal-
ities.
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
2
3.2. Ophthalmological ndings
At the anamnestic evaluation, the subject referred night-blindness in
the last year. BCVA was 20/120 in both eyes. Fundus examination re-
vealed a normal optic disk, punctiform lens opacities, dystrophy of
Retinal Pigmented Epithelium (RPE) with bone spicule-shaped pigment
deposits arranged within and beyond the vascular arcades with a
normal appearance of the far periphery and macular dystrophy
(Fig. 1a). GVF showed a constricted visual eld. Specically, the
average radius was 17° and 27° using the III4e target stimulus size and
30° and 40° using the V4e target stimulus size in the right and left eye,
respectively. OCT scans showed RPE dystrophy with loss of the EZ band
in both eyes and revealed multiple inner retinal cystoid spaces (with a
mean macular thickness of 260 μm and 293 μm in the right and left eye,
respectively) (Fig. 1b). FAF imaging revealed hyper- and hypo-auto-
uorescent dots in the posterior pole and beyond the temporal vascular
arcades sparing the fovea in both eyes. Dark-adapted 0.01 ERG re-
sponses were below noise level, whereas dark-adapted 3.0 responses
were subnormal in both eyes with a b/a ratio < 1. Light-adapted ERG
responses were subnormal in both eyes. Given the presence of cystoid
spaces, the patient was prescribed treatment with oral acetazolamide
(250 mg/die) and was regularly followed up. Over the two-year follow-
up period, we recorded a reduction of cystic spaces (with mean macular
thickness of 185 μm and 208 μm in the right and left eye, respectively)
without signicant changes in BCVA and in GVF (Fig. 1b). Taken to-
gether, the ophthalmological ndings conrmed an ocular phenotype
compatible with RP complicated by macular cystoid edema.
OCT scans over a two-year follow-up showed an improvement of the
macular edema in the patient following treatment with acetazolamide.
3.3. Audiological ndings
There was no history of sensorineural deafness on either side of the
family. The patient has one younger sister with a normal audiometric
threshold. Parents reported that the child underwent the universal
newborn hearing screening program (performed by law in Italy)
twenty-four hours after birth with normal results (a document reporting
the results of TEOAE was not available). At 3 months of age, the general
practitioner suspected hearing loss and recommended a specic
audiological evaluation which was performed by ABR. This test, con-
sidered as the gold standard for the early diagnosis of childhood deaf-
ness, showed an abnormal morphology and latency of the auditory
brain response waves. The latency of the I, III and V waves was in-
creased and only the V wave was observed up to 60 dB hearing level
bilaterally. This nding suggested an auditory decit characterized by
severe SNHL in both ears.
The impedance test showed a Type Atympanogram bilaterally,
considered normal, and the presence of the stapedial reexes (SR) in
ipsi and contra in the entire frequency range. The Metz recruitment test,
i.e. the gap between the acoustic reex threshold and the pure-tone
audiometry hearing threshold level, was positive and indicated a co-
chlear SNHL. Subsequent checks performed at the age of 6 and
9 months by ABR conrmed the SNHL diagnosis prompting for an early
rehabilitation treatment by digital hearing aids and speech therapy that
started at 10 months of age. The patient has been regularly undergoing
an annual audiological check from 2008 to date. The most recent ones,
performed at the age of 9 and 10, conrmed the presence of bilateral
and symmetric SNHL of a severe degree (according to World Health
Organization - Grades of Hearing Impairment) that does not appear to
be progressive over time, with a sloping audiogramand a PTA
(without hearing device) around 70 dB HL (Fig. 2). Further annual
follow-ups are needed to monitor the hearing impairment over time.
Fig. 1. Fundus imaging (a) and OCT scans (b) of the reported case.
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
3
3.4. Phoniatric and neuropsychological ndings
A phoniatric and neuropsychological assessment was performed as
learning disabilities were referred by the patient's teacher and parents.
The administration of ad hoc tests revealed a decit of language skills
both in input and output. The obtained results were below the average
expected for this age (< 2 SD) and indicated a global immaturity of
speech abilities as well as inadequate capacities of phonological dis-
crimination, lexical and grammar understanding.
Phonological and metaphonological skills (particularly the analy-
tical ones) as well as the verbal comprehension were severely com-
promised compared to the other children in the same age-group This
suggests that the child had learning disabilities, especially with regards
to reading and writing. Working memory abilities and short-term verbal
memory abilities were compromised compared to average degrees. For
this reason, a further clinical examination was recommended in order
to evaluate the presence of an intellectual disability.
Neuropsychological evaluation, according to the results of beha-
vioral observation and clinical interviews, showed that the patient was
aected by Separation anxiety (revealed by SCARED score) that im-
pacts on school performance. The scores obtained by the administration
of the WISC-IV (IQ total score = 65) underline the presence of a mild
intellectual disability. In light of these ndings, the learning disabilities
observed in the patient (i.e. in writing and reading) can be considered
secondary to the cognitive delay.
Magnetic resonance imaging (MRI) of the brain did not reveal any
signs of leukodystrophy or other signicant brain abnormalities.
3.5. Molecular analysis identifying biallelic variants in PEX1
The genetic analysis revealed the presence of two variants of un-
certain signicance (VUS) in PEX1 (NM_000466). The identied var-
iants were present in heterozygosity in the proband and their frequency
in population databases (e.g. gnomAD, the ExAC database, the 1000
Genomes project) was compatible with a possible pathogenic role. The
missense variant c.274G > C; p.(Val92Leu) is predicted to interfere
with a canonical splice site (cadd-13 score = 24.6) and was reported in
a homozygous state in a patient with atypical Zellweger syndrome [20].
The second variant c.2140_2145dup; p.(Ser714_Gln715dup) is a novel
non-frameshift duplication of six nucleotides predicted to introduce a
third copy of a Ser-Gln repeat at amino acid position 715 (UniProtKB:
O43933). Both variants were validated by Sanger sequencing in the
proband and segregated properly in the unaected parents, conrming
their presence in trans in the patient (i.e. p.(Val92Leu) on the paternal
allele and p.(Ser714_Gln715dup) on the maternal one), consistent with
the recessive pattern of inheritance of PEX1 variants (Fig. 3).
PEX1 is responsible for ZSD-PBD and HS. Therefore, the identica-
tion of these variants in PEX1 prompted us to reexamine the patient's
phenotype in search for clinical features that may have been missed at
rst evaluation.
Sanger sequencing traces indicate compound heterozygosity of the
two variants in exon 3 and exon 13 in the patient (II:1) and hetero-
zygosity in the unaected parents (I:1, I:2). In exon 13, the duplicated
bases of the c.2140_2145dup variant are connected with a dashed line
to the corresponding peaks in the proband's chromatogram. Arrow in-
dicates the proband. Ref; Reference sequence.
3.6. Laboratory analysis and VLCFA assay
Routine serum VLCFA and branched-chain fatty acid (phytanic and
pristanic acid) measurements were performed to check for defects in
the metabolism of fatty acids, given the role of peroxins in this process.
The proband was not on special diet and fasting serum samples were
collected. The majority of the values were within the normal range,
with C26:0 slight increase (Table 1)[
21].
VLCFA plasma concentration may vary, with subjects demonstrating
normal or only modest increases, especially in milder or atypical forms
of PBDs [22,23]; moreover normal or slightly elevated results of routine
serum VLFCA and phytanic acid in patients aected by mild PBDs have
already been described by other authors [20,24,25].
Anyway we have to consider that the value of C26:0 obtained in our
patient can be due to nonspecic factors and, for this, further im-
munocytochemical studies in cultured broblasts (considered a more
sensitive indicator for a mild PBD) would be performed in the future as
well as measuring plasma pipecolic acid and the bile acid intermediates
dihydroxycholestanoic (DHCA) and trihydroxycholestanoic (THCA)
may be helpful.
Fig. 2. Pure tone audiometry (air and bone conduction) in the right ear (in red)
and in the left ear (in blue) in the frequency range between 250 Hz - 8 KHz.
Down-sloping audiogram with severe bilateral sensorineural hearing loss is
more evident in the higher frequencies.
Fig. 3. Biallelic variants identied in PEX1.
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
4
3.7. Dental ndings
At the extraoral examination, on the frontal plane, the patient
showed a symmetrical face both on transverse and vertical planes, with
competent lips. At sagittal examination, the prole was straight. The
intraoral examination revealed a mixed dentition, as expected at this
age [26]. In particular, the upper and lower permanent incisors and the
rst permanent molar were present. The remaining dentition consisted
of primary teeth (canines and molars), except for the two rst primary
inferior molars, which were exfoliated (as reported by the patient) and
replaced by barely visible, thin, erupting cusps. Several enamel and
structural defects of various degree aected the permanent teeth [27]
(Fig. 4a).
Specically, the upper central incisors displayed white spots on the
vestibular surfaces. Moreover, the occlusal areas of the rst permanent
molars and the mesial-vestibular margin of the inferior right second
incisor were aected by enamel hypoplasia. These appeared yellowish
and had an irregular shape at the cusps. The erupting rst lower pre-
molars also seemed to have similar defects.
The X-ray orthopantomography (Fig. 4b) revealed no agenesis, since
all the permanent teeth were present at a variable degree of maturation
and root formation, including the four buds of the third molars. The
anatomy of the roots of the permanent teeth did not show any obvious
abnormalities. However, the enamel density of canines and premolars
appeared reduced and their cusps were slightly hypoplastic. The pulp
chambers were dimensionally enlarged as expected at that age, yet not
excessively big as in taurodontic teeth, as previously reported [28,29].
On the vertical plane, the occlusion was altered by the presence of an
anterior open bite due to mouth breathing reported by the patient. The
same behavior was responsible for the discrepancies observed on the
transversal plane, between maxilla and mandible, namely a reduced
bilateral overjet and a high-arched palate. Oral mucosae were healthy
and normochromic. Tongue and teeth were covered by a visible layer of
dental plaque, which was subsequently removed by professional oral
hygienist.
Overall, the structural and chromatic dental defects pointed towards
the diagnosis of enamel defects with hypomineralization, particularly
aecting incisors and molars.
3.8. Dermatological ndings
Total body skin examination revealed a true leukonychia partialis
equally aecting all ngernails and minimally sparing the distal edges
of each nail. Leukonychia was conrmed by onychoscopy showing a
whitish discoloration of nail plates which did not disappear after
pressure on the nail matrix (Fig. 5a). Moreover, a careful examination
of scalp hair showed an alternation of light and dark bands of hair
shafts. This was conrmed by trichoscopy and corresponded to ab-
normal cavities in the cortex of the hair shaft compatible with the di-
agnosis of pili annulati (Fig. 5b).
4. Discussion
In this report, we employed a multidisciplinary approach to char-
acterize in detail the clinical phenotype of a pediatric patient with a
Table 1
Main results of serum standard biochemical markers in the patient.
Serum VLCFA Patient Normal control range
C22:0 57.9 μmol/L 26.575.3 μmol/L
C24:0 44.9 μmol/L 24.973.0 μmol/L
C26:0 1.130 μmol/L
a
0.4600.960 μmol/L
C24:0/C22:0 0.77 0.621.01
C26:0/C22:0 0.019 0.0080.026
Pristanic acid 0.13 μmol/L Traces - 1.50 μmol/L
Phytanic acid 1.76 μmol/L Traces - 7.0 μmol/L
Pristanic/Phytanic Ratio 0.07 0.010.60 μmol/L
a
Out-of-normal-range value.
Fig. 4. Main dental ndings of the reported case.
a) High-arched palate and visible enamel hypoplasia of the rst permanent
upper molars; further chromatic and structural enamel alterations of the rst
permanent lower molars b) Orthopantomography reveal the presence of all the
permanent teeth expected, lack of taurodontism, nor agenesis. The cusps of the
permanent canines and premolars, particularly of the mandible, seem thinner
and, presumably hypoplastic.
Fig. 5. Dermatological ndings of the reported case.
a) Leukonychia partialis of all ngernails sparing the distal margin of the nail
plates, better magnied by onychoscopy in the right caption b) Clinically de-
tectable banding of hair, conrmed by trichoscopy and light microscopy,
showing abnormal cavities in the cortex of the hair shaft typical of pili annulati.
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
5
mild form of PBD-ZSD.
The patient was referred to our clinic with an apparently isolated
pre-verbal SNHL and night blindness. The full ophthalmological eva-
luation revealed a pericentral form of RP complicated by bilateral
macular edema. The ocular and audiological phenotype along with the
absence of dysmorphic features pointed towards a preliminary clinical
diagnosis of Usher syndrome. However, the patient's phenotype of pre-
puberal RP and early onset, severe SNHL did not t well in the spectrum
of Usher disease. This was because in Usher type 1 the presence of pre-
puberal RP is associated with congenital SNHL of a profound degree,
whereas in Usher type 2 a moderate-to-severe (generally congenital)
SNHL is normally associated with post-puberal RP.
In parallel, genetic testing by clinical exome sequencing did not
reveal variants in genes that cause Usher syndrome, but identied
biallelic variants in PEX1, a gene responsible for peroxisome biogenesis
disorders. The two identied variants were classied as VUS according
to the American College of Medical Genetics and Genomics (ACMG)
guidelines. The p.(Ser714_Gln715dup) variant is novel and absent from
reference databases. Its translation introduces a third copy of a Ser-Gln
pair in a small repeat region of PEX1. The second mutation is a missense
variant (p.(Val92Leu)) predicted in silico to interfere with a canonical
acceptor splice site. We believe that both variants are likely to be pa-
thogenic since they were found in trans (as conrmed by segregation
analysis) in a patient presenting a clinical phenotype compatible with a
peroxisome biogenesis disorder. Future in vitro studies should experi-
mentally verify the functional consequences of the identied variants.
In support of its potential causative role, the variant p.(Val92Leu) has
been previously described in homozygous state in a Turkish patient
with a non-classical ZS, although it was not specied whether zygosity
was conrmed by segregation in the parents [20]. This patient, contrary
to the case reported herein, presented several dysmorphic features (e.g.,
large fontanelle, wide sutures, high forehead, broad nasal bridge, ex-
ternal ear deformity and sickle foot), hypotonia, severe psychomotor
retardation but no ocular ndings [20]. A possible explanation for the
absence of ocular ndings could be attributed to the patient's age at
examination (i.e. 2 years old), also considering that our case reported
the rst symptoms of visual impairment at the age of 8 years.
The similarity between Usher syndrome and mild phenotypes of
PBD-ZSD has been previously reported [21,3032]. We, therefore,
pursued a comprehensive clinical assessment to dierentiate between
the two possibilities. Neuropsychological assessment of the patient re-
vealed the presence of a mild cognitive impairment, a common feature
in the spectrum of peroxisome disorders. Brain MRI did not reveal
generalized defects. Moreover, the patient had a lower weight com-
pared with normal population associated with a signicant food se-
lectivity and poor feeding, features that have been described in inter-
mediate-mild forms of ZSD [1,5]. Oral examination showed minor oral
abnormalities, in terms of enamel defects, as previously described in
cases of intermediate-milder PBD-ZSD [5]. The dermatological ndings
present in the patient (i.e. leukonychia, Beau's lines and hair defects,)
are not considered as diagnostic criteria in the spectrum of PBD-ZSD.
Nevertheless, mutations in PEX1 and PEX6 genes may determine nail
and hair defects [31]. Taking into account both the clinical and genetic
ndings, we formulated a diagnosis of a mild PBD-ZSD. Plasmatic
VLCFA levels were overall normal, except for a slight increase of C26:0.
This was in line with previous studies showing that individuals with
very mild/mild PBDs do not necessarily demonstrate signicantly al-
tered values in VLCFA metabolic screening tests as observed for patients
with severe forms of ZSD [3335]. Unfortunately, measurement of
C26:0-lysoPC, which was recently proposed as a novel, sensitive serum
biomarker for the diagnosis of mild PBD-ZSD [1], could not be per-
formed in the patient, as well as skin biopsy and immunocytochemical
studies in cultured broblasts as mentioned above.
5. Conclusions
In conclusion, we report here a case of mild form of PBD-ZSD
characterized by slight abnormalities of VLCFA, early onset SNHL,
atypical RP, enamel defects, nail abnormalities, minor feeding problems
and mild cognitive impairment in absence of hypotonia, dysmorphic
features and other major abnormalities. The association of hearing loss,
enamel defect and nail abnormalities, with or without macular dys-
trophy, has been described also in Heimler syndrome (HS), which is
considered the mildest form known to date in the spectrum of PBD
[1215]. Although these clinical features are observed in our patient,
the presence of a mild cognitive impairment and feeding problems
renders her phenotype more compatible with a mild form of ZSD [1]
which has a slightly more severe clinical presentation compared to the
typical Heimler syndrome [1316,22,31].
Based on the presented case, we recommend that patients with vi-
sual and hearing impairment perform a comprehensive mutation
screening that includes the PEX genes.The molecular analysis can be
instrumental for the early identication of patients with mild forms of
PBD-ZSD that may overlap with other syndromic diseases. Moreover,
the timely and proper diagnosis of these rare cases requires the com-
bined eort of a multidisciplinary team of clinicians. Following initial
diagnosis, periodic multidisciplinary follow-ups are required to monitor
disease progression and ensure appropriate disease management.
Declaration of Competing Interest
None of the authors has a conict of interest.
Acknowledgement
This work was supported by grant from the Italian Fondazione
Roma (to S.B. and F.S.). This study was also funded by Ministero
dell'Istruzione, dell'Università e della Ricerca (MIUR) under PRIN 2015
(to S.B. and F.S.)
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://
doi.org/10.1016/j.ymgmr.2020.100615.
References
[1] F.C. Klouwer, K. Berendse, S. Ferdinandusse, R.J. Wanders, M. Engelen, B.T. Poll-
The, Zellweger spectrum disorders: clinical overview and management approach,
Orphanet. J. Rare Dis. 10 (2015) 151.
[2] A. Schluter, S. Fourcade, E. Domenech-Estevez, et al., PeroxisomeDB: a database for
the peroxisomal proteome, functional genomics and disease, Nucleic Acids Res. 35
(2007) D815D822.
[3] S.J. Steinberg, G. Dodt, G.V. Raymond, N.E. Braverman, A.B. Moser, H.W. Moser,
Peroxisome biogenesis disorders, Biochim. Biophys. Acta 1763 (2006) 17331748.
[4] N.E. Braverman, M.D. DAgostino, G.E. Maclean, Peroxisome biogenesis disorders:
biological, clinical and pathophysiological perspectives, Dev. Disabil. Res. Rev. 17
(2013) 187196.
[5] N.E. Braverman, G.V. Raymond, W.B. Rizzo, et al., Peroxisome biogenesis disorders
in the Zellweger spectrum: an overview of current diagnosis, clinical manifesta-
tions, and treatment guidelines, Mol. Genet. Metab. 117 (2016) 313321.
[6] G.N. Wilson, R.G. Holmes, J. Custer, et al., Zellweger syndrome: diagnostic assays,
syndrome delineation, and potential therapy, Am. J. Med. Genet. 24 (1986) 6982.
[7] N. Shimozawa, T. Tsukamoto, Y. Suzuki, et al., A human gene responsible for
Zellweger syndrome that aects peroxisome assembly, Science 255 (1992)
11321134.
[8] N. Preuss, U. Brosius, M. Biermanns, A.C. Muntau, E. Conzelmann, J. Gartner, PEX1
mutations in complementation group 1 of Zellweger spectrum patients correlate
with severity of disease, Pediatr. Res. 51 (2002) 706714.
[9] S. Steinberg, L. Chen, L. Wei, et al., The PEX gene screen: molecular diagnosis of
peroxisome biogenesis disorders in the Zellweger syndrome spectrum, Mol. Genet.
Metab. 83 (2004) 252263.
[10] B.T. Poll-The, J. Gootjes, M. Duran, et al., Peroxisome biogenesis disorders with
prolonged survival: phenotypic expression in a cohort of 31 patients, Am. J. Med.
Genet. A 126A (2004) 333338.
[11] C. Walter, J. Gootjes, P.A. Mooijer, et al., Disorders of peroxisome biogenesis due to
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
6
mutations in PEX1: phenotypes and PEX1 protein levels, Am. J. Hum. Genet. 69
(2001) 3548.
[12] A. Heimler, J.E. Fox, J.E. Hershey, P. Crespi, Sensorineural hearing loss, enamel
hypoplasia, and nail abnormalities in sibs, Am. J. Med. Genet. 39 (1991) 192195.
[13] K.R. Ong, S. Visram, S. McKaig, L.A. Brueton, Sensorineural deafness, enamel ab-
normalities and nail abnormalities: a case report of Heimler syndrome in identical
twin girls, Eur. J. Med. Genet. 49 (2006) 187193.
[14] C. Pollak, M. Floy, B. Say, Sensorineural hearing loss and enamel hypoplasia with
subtle nail ndings: another family with Heimlers syndrome, Clin. Dysmorphol. 12
(2003) 5558.
[15] M. Tischkowitz, C. Clenaghan, S. Davies, L. Hunter, J. Potts, S. Verhoef,
Amelogenesis imperfecta, sensorineural hearing loss, and Beaus lines, a second case
report of Heimlers syndrome, J. Med. Genet. 36 (1999) 941943.
[16] L.H. Lima, I.A. Barbazetto, R. Chen, L.A. Yannuzzi, S.H. Tsang, R.F. Spaide, Macular
dystrophy in Heimler syndrome, Ophthalmic Genet. 32 (2011) 97100.
[17] A.B. Moser, N. Kreiter, L. Bezman, et al., Plasma very long chain fatty acids in 3,000
peroxisome disease patients and 29,000 controls, Ann. Neurol. 45 (1999) 100110.
[18] T.J. Stradomska, A. Tylki-Szymanska, Examination of very long chain fatty acids in
diagnosis of x-linked adrenoleukodystrophy, Pediatr. Pol. 71 (1996) 197201.
[19] V. Di Iorio, M. Karali, R. Brunetti-Pierri, et al., Clinical and genetic evaluation of a
cohort of pediatric patients with severe inherited retinal dystrophies, Genes 8
(2017).
[20] H. Rosewich, A. Ohlenbusch, J. Gartner, Genetic and clinical aspects of Zellweger
spectrum patients with PEX1 mutations, J. Med. Genet. 42 (2005) e58.
[21] M.J. Ventura, D. Wheaton, M. Xu, et al., Diagnosis of a mild peroxisomal phenotype
with next-generation sequencing, Mol. Genet. Metab. Rep. 9 (2016) 7578.
[22] I. Ratbi, I.C. Jaouad, H. Elorch, et al., Severe early onset retinitis pigmentosa in a
Moroccan patient with Heimler syndrome due to novel homozygous mutation of
PEX1 gene, Eur. J. Med. Genet. 59 (2016) 507511.
[23] R.J. Wanders, H.R. Waterham, Peroxisomal disorders I: biochemistry and genetics
of peroxisome biogenesis disorders, Clin. Genet. 67 (2005) 107133.
[24] P. Lipinski, P. Stawinski, M. Rydzanicz, et al., Mild Zellweger syndrome due to
functionally conrmed novel PEX1 variants, J. Appl. Genet. 61 (2020) 8791.
[25] C.E. Smith, J.A. Poulter, A.V. Levin, et al., Spectrum of PEX1 and PEX6 variants in
Heimler syndrome, Eur. J. Hum. Genet. 24 (2016) 15651571.
[26] E. Gentile, D. Di Stasio, R. Santoro, et al., In vivo microstructural analysis of enamel
in permanent and deciduous teeth, Ultrastruct. Pathol. 39 (2015) 131134.
[27] M. Contaldo, D. Di Stasio, R. Santoro, et al., Non-invasive in vivo visualization of
enamel defects by reectance confocal microscopy (RCM), Odontology 103 (2015)
177184.
[28] V.A. de la Pena, M.C. Valea, Treatment of enamel hypoplasia in a patient with usher
syndrome, J. Am. Dent. Assoc. 142 (2011) 938941.
[29] M. Contaldo, R. Serpico, A. Lucchese, In vivo imaging of enamel by reectance
confocal microscopy (RCM): non-invasive analysis of dental surface, Odontology
102 (2014) 325329.
[30] A. Raas-Rothschild, R.J. Wanders, P.A. Mooijer, et al., A PEX6-defective perox-
isomal biogenesis disorder with severe phenotype in an infant, versus mild phe-
notype resembling usher syndrome in the aected parents, Am. J. Hum. Genet. 70
(2002) 10621068.
[31] I. Ratbi, K.D. Falkenberg, M. Sommen, et al., Heimler syndrome is caused by
Hypomorphic mutations in the peroxisome-biogenesis genes PEX1 and PEX6, Am. J.
Hum. Genet. 97 (2015) 535545.
[32] M.S. Zaki, R. Heller, M. Thoenes, et al., PEX6 is expressed in photoreceptor cilia and
mutated in Deafblindness with enamel dysplasia and microcephaly, Hum. Mutat. 37
(2016) 170174.
[33] M. Rydzanicz, T.J. Stradomska, E. Jurkiewicz, et al., Mild Zellweger syndrome due
to a novel PEX6 mutation: correlation between clinical phenotype and in silico
prediction of variant pathogenicity, J. Appl. Genet. 58 (2017) 475480.
[34] A. Zeharia, M.S. Ebberink, R.J. Wanders, et al., A novel PEX12 mutation identied
as the cause of a peroxisomal biogenesis disorder with mild clinical phenotype, mild
biochemical abnormalities in broblasts and a mosaic catalase immunouorescence
pattern, even at 40 C, J. Hum. Genet. 52 (2007) 599.
[35] L. Regal, M.S. Ebberink, N. Goemans, et al., Mutations in PEX10 are a cause of
autosomal recessive ataxia, Ann. Neurol. 68 (2010) 259263.
M.R. Barillari, et al. Molecular Genetics and Metabolism Reports 24 (2020) 100615
7
... [1] Survival beyond the first year of life is rare due to the array of symptoms and organ involvement in severe ZS. [2] The milder phenotypes may have a better prognosis and present into adulthood with associated defects such as sensorineural hearing loss, retinal diseases, leukodystrophy, and cognitive delay. [5,6] UHS or spun glass hair is a rare structural anomaly of the hair shaft. [7] Both sporadic and inherited forms are described. ...
... Previously reported cases of ZS with PEX 1 and PEX 6 gene mutations had dermatological findings of Beau's lines, leukonychia, and hair abnormalities like pili annulati. [5] Another case report diagnosed with PEX 12 mutation and milder ZS, had cognitive impairment, visual and hearing loss. [5] In our case, the patient has liver and hair abnormalities (UHS) alone. ...
... [5] Another case report diagnosed with PEX 12 mutation and milder ZS, had cognitive impairment, visual and hearing loss. [5] In our case, the patient has liver and hair abnormalities (UHS) alone. The definitive diagnosis of ZS may be achieved by genetic testing, which was done in this case. ...
Article
Full-text available
Zellweger syndrome (ZS) is a rare autosomal recessive, peroxisomal biogenesis disorder (PBD) that occurs due to a mutation in any of the thirteen peroxin (PEX) genes. It is reported to manifest with varying degrees of severity, ranging from non-specific gastrointestinal abnormalities, nail and enamel defects to multisystem involvement (cerebro-hepato-renal syndrome, eye, ear, and neurological abnormalities). Uncombable hair syndrome (UHS) is a rare hair shaft disorder characterized by dry, frizzy, unmanageable hair. Diagnosis of UHS can be confirmed by scanning electron microscopy (SEM), which reveals a triangular cross-section of the hair. We report a case of UHS with a hitherto unreported association of ZS (due to a homozygous mutation of PEX 12).
... As a result, many patients may be misdiagnosed with Usher syndrome. 2 Several reports exist of patients with hearing loss and retinopathy who showed negative results for pathogenic variants in genes associated with Usher syndrome and later were discovered to have a PBD. 2,3 Dysfunction of a dynamic, single membrane-bound organelle called the peroxisome underlies the PBDs. ...
... 2 Several reports exist of patients with hearing loss and retinopathy who showed negative results for pathogenic variants in genes associated with Usher syndrome and later were discovered to have a PBD. 2,3 Dysfunction of a dynamic, single membrane-bound organelle called the peroxisome underlies the PBDs. Peroxisomes perform diverse functions, including b-oxidation of very long-chain fatty acids, a-oxidation of branchedchain fatty acids, bile acid and plasmalogen synthesis, and the detoxification of reactive oxygen species. ...
... Pathogenic variants in PEX genes recently were identified in other patients with a clinical diagnosis of Usher syndrome, but genetic testing was not revealing. 2 Compound heterozygous mutations in PEX6 (NM_000287.4) were discovered: c.802_815del:p.(Asp268 Cysfs*8) and c.35T/C: p.(Phe12Ser). The deletion was determined to be inherited paternally and the missense mutation was determined to be inherited maternally. ...
Article
Full-text available
Purpose Peroxisomal biogenesis disorders (PBDs) represent a spectrum of recessive conditions that result in vision loss, sensorineural hearing loss, neurologic dysfunction, and other abnormalities due to aberrant peroxisomal function caused by mutations in PEX genes. With no treatments currently available, we sought to investigate the disease mechanism in a patient with a PBD caused by defects in PEX6 and to probe whether over-expression of PEX6 could restore peroxisome function and potentially offer therapeutic benefit. Design Laboratory-based study. Participants A 12-year-old male presented to our clinic with hearing loss and retinopathy. After a negative Usher syndrome panel, targeted genetic testing revealed compound heterozygous mutations in PEX6. These included a 14-nucleotide deletion (c.802_815del: p.(Asp268Cysfs*8)) and a milder missense variant (c.35T>C:(p.Phe12Ser)). Methods Patient-derived skin fibroblasts were cultured, and a PEX6 knock-out cell line was developed using CRISPR/Cas9 technology in HEK293T cells to emulate a more severe disease phenotype. Immunoblot analysis of whole cell lysates was performed to assess peroxisome number. Immunofluorescence studies used antibodies against components of the peroxisomal protein import pathway to interrogate the effects of mutations in PEX6 on protein trafficking. Main Outcome Measures Primary outcome measures were peroxisome abundance and matrix protein import. Results Peroxisome number was not significantly different between control fibroblasts and patient fibroblasts; however, fewer peroxisomes were observed in PEX6 knock-out cells compared to wild-type (P = 0.04). Analysis by immunofluorescent microscopy showed significantly impaired PTS1- and PTS2-mediated matrix protein import in both patient fibroblasts and PEX6 knock-out cells. Over-expressing PEX6 resulted in improved matrix protein import in PEX6 knock-out cells. Conclusions Mutations in PEX6 were found to be responsible for the combined hearing loss and retinopathy observed in our patient. The primary peroxisomal defect in our patient’sskin fibroblasts was impaired peroxisomal protein import as opposed to a reduction in the number of peroxisomes. Genetic strategies that introduce wild-type PEX6 into cells deficient in PEX6 protein show promise in restoring peroxisome function. Future studies on patient-specific iPSC-derived RPE cells may further clarify the role of PEX6 in the retina and the potential for gene therapy as a treatment approach in these patients.
... secondary cone damage) [31]. Furthermore, the diverse retinal pathology is underscored by the fact that several patients presenting with a disease phenotype of Leber congenital amaurosis [32][33][34] and Usher syndrome [20,[35][36][37], two well-known diseases that are characterized by pigmentary retinopathy, have been re-diagnosed as suffering from ZSD. ...
... The retinal structure of two mild PEX1 patients was described. These patients were initially suspected to suffer from Usher syndrome [37] and Leber congenital amaurosis [33]. The alleged Usher syndrome patient developed neurosensory hearing loss at the age of 3 months and night-blindness at the age of 9 years. ...
... Fundoscopy and OCT revealed RPE and macular dystrophy complicated by macular cystoid oedema. Dark-adapted ERG responses were strongly impaired while light-adapted ERG was only mildly affected [37]. The retinal pathology of the re-diagnosed Leber congenital amaurosis patient appeared more complicated, as the macula and periphery were affected, while the perifoveal region was not. ...
Article
Full-text available
Peroxisomes are multifunctional organelles, well known for their role in cellular lipid homeostasis. Their importance is highlighted by the life-threatening diseases caused by peroxisomal dysfunction. Importantly, most patients suffering from peroxisomal biogenesis disorders, even those with a milder disease course, present with a number of ocular symptoms, including retinopathy. Patients with a selective defect in either peroxisomal α- or β-oxidation or ether lipid synthesis also suffer from vision problems. In this review, we thoroughly discuss the ophthalmological pathology in peroxisomal disorder patients and, where possible, the corresponding animal models, with a special emphasis on the retina. In addition, we attempt to link the observed retinal phenotype to the underlying biochemical alterations. It appears that the retinal pathology is highly variable and the lack of histopathological descriptions in patients hampers the translation of the findings in the mouse models. Furthermore, it becomes clear that there are still large gaps in the current knowledge on the contribution of the different metabolic disturbances to the retinopathy, but branched chain fatty acid accumulation and impaired retinal PUFA homeostasis are likely important factors.
... Peroxisomal Biogenesis and Zellweger Spectrum Disorders (PBD-ZSD) are a group of rare autosomal recessive disorders caused by mutations in PEX genes, characterized by defective peroxisome assembly and function. Patients with PBD-ZSD display physiological, developmental, and neurological complications that include visual and sensorineural hearing loss (SNHL) [1][2][3]. Based on genotype-phenotype correlations, PBD-ZSD are classified as severe, intermediate, or mild within the PBD-ZSD spectrum [4]. Severe forms of the disease are associated with a complete loss of peroxisomal function. ...
Article
Full-text available
Peroxisome Biogenesis Disorders (PBD) and Zellweger syndrome spectrum disorders (ZSD) are rare genetic multisystem disorders that include hearing impairment and are associated with defects in peroxisome assembly, function, or both. Mutations in 13 peroxin (PEX) genes have been found to cause PBD-ZSD with ~70% of patients harboring mutations in PEX1. Limited research has focused on the impact of peroxisomal disorders on auditory function. As sensory hair cells are particularly vulnerable to metabolic changes, we hypothesize that mutations in PEX1 lead to oxidative stress affecting hair cells of the inner ear, subsequently resulting in hair cell degeneration and hearing loss. Global deletion of the Pex1 gene is neonatal lethal in mice, impairing any postnatal studies. To overcome this limitation, we created conditional knockout mice (cKO) using Gfi1Creor VGlut3Cre expressing mice crossed to floxed Pex1 mice to allow for selective deletion of Pex1 in the hair cells of the inner ear. We find that Pex1 excision in inner hair cells (IHCs) leads to progressive hearing loss associated with significant decrease in auditory brainstem responses (ABR), specifically ABR wave I amplitude, indicative of synaptic defects. Analysis of IHC synapses in cKO mice reveals a decrease in ribbon synapse volume and functional alterations in exocytosis. Concomitantly, we observe a decrease in peroxisomal number, indicative of oxidative stress imbalance. Taken together, these results suggest a critical function of Pex1 in development and maturation of IHC-spiral ganglion synapses and auditory function.
... In the past years, the implementation of next-generation sequencing (NGS) in the clinical setting has allowed the identification of an increasing number of mild clinical conditions in adult patients who frequently present normal or slight biochemical alterations in plasma [5,[12][13][14][15][16][17][18][19]. To this regard, Heimler Syndrome (HS) has been recognized as a peroxisome biogenesis disorder within the ZSDs and added to the (very) mild end of the clinical spectrum. ...
Article
Full-text available
Peroxisomal biogenesis disorders (PBDs) are a heterogeneous group of genetic diseases. Multiple peroxisomal pathways are impaired, and very long chain fatty acids (VLCFA) are the first line biomarkers for the diagnosis. The clinical presentation of PBDs may range from severe, lethal multisystemic disorders to milder, late-onset disease. The vast majority of PBDs belong to Zellweger Spectrum Disordes (ZSDs) and represents a continuum of overlapping clinical symptoms, with Zellweger syndrome being the most severe and Heimler syndrome the less severe disease. Mild clinical conditions frequently present normal or slight biochemical alterations, making the diagnosis of these patients challenging. In the present study we used a combined WES and RNA-seq strategy to diagnose a patient presenting with retinal dystrophy as the main clinical symptom. Results showed the patient was compound heterozygous for mutations in PEX1. VLCFA were normal, but retrospective analysis of lysosphosphatidylcholines (LPC) containing C22:0–C26:0 species was altered. This simple test could avoid the diagnostic odyssey of patients with mild phenotype, such as the individual described here, who was diagnosed very late in adult life. We provide functional data in cell line models that may explain the mild phenotype of the patient by demonstrating the hypomorphic nature of a deep intronic variant altering PEX1 mRNA processing.
... Various syndromic genetic disorders can be associated with oral manifestations, and, while some of them are extremely rare [23][24][25][26][27], many others are commonly seen: this is the case with Down syndrome (DS). DS is well known and its phenotypic aspect has been extensively described, but there is no systematic report on the particular prevalence of oral mucosal manifestation in children with DS. ...
Article
Full-text available
Down syndrome (DS) is an autosomal disorder associated with mental and physical involvement. The typical craniofacial phenotype and the dental anomalies in DS subjects have been widely described, but a systematic report on the manifestations affecting the oral mucosae in children with DS is still lacking. This systematic review aimed to establish the prevalence of oral mucosal manifestations in children/young adults with DS. Pubmed, Web of Science, and Scopus were investigated in September 2020. Documents in English on DS children/young adults (up to 25 years) reporting oral mucosal findings were considered. Study quality was assessed with ROBIN-I. Of the 150 references retrieved, 14 studies were considered eligible. The risk of bias ranged from low to unclear. Fissured tongue appeared to increase with age and was more prevalent in DS children than in the general population. Lip fissures and cheilitis were heterogeneously reported. Candida spp. carriage with and without active candidiasis was more frequent in DS children/young adults than in controls. C. albicans was the most prevalent species. Few other oral mucosal conditions have been reported sporadically. The heterogeneity of the works revealed the need for more appropriate oral examination to intercept the oral manifestations of oral mucosa and prevent recurrent candidiasis.
Article
Purpose: This cross-sectional study describes the ophthalmological and general phenotype of 10 patients from six different families with a comparatively mild form of Zellweger spectrum disorder (ZSD), a rare peroxisomal disorder. Methods: Ophthalmological assessment included best-corrected visual acuity (BCVA), perimetry, microperimetry, ophthalmoscopy, fundus photography, spectral-domain optical coherence tomography (SD-OCT), and fundus autofluorescence (FAF) imaging. Medical records were reviewed for medical history and systemic manifestations of ZSD. Results: Nine patients were homozygous for c.2528 G > A (p.Gly843Asp) variants in PEX1 and one patient was compound heterozygous for c.2528 G>A (p.Gly843Asp) and c.2097_2098insT (p.Ile700TyrfsTer42) in PEX1. Median age was 22.6 years (interquartile range (IQR): 15.9 - 29.9 years) at the most recent examination, with a median symptom duration of 22.1 years. Symptom onset was variable with presentations of hearing loss (n = 7) or nyctalopia/reduced visual acuity (n = 3) at a median age of 6 months (IQR: 1.9-8.3 months). BCVA (median of 0.8 logMAR; IQR: 0.6-0.9 logMAR) remained stable over 10.8 years and all patients were hyperopic. Fundus examination revealed a variable retinitis pigmentosa (RP)-like phenotype with rounded hyperpigmentations as most prominent feature in six out of nine patients. Electroretinography, visual field measurements, and microperimetry further established the RP-like phenotype. Multimodal imaging revealed significant intraretinal fluid cavities on SD-OCT and a remarkable pattern of hyperautofluorescent abnormalities on FAF in all patients. Conclusion: This study highlights the ophthalmological phenotype resembling RP with moderate to severe visual impairment in patients with mild ZSD. These findings can aid ophthalmologists in diagnosing, counselling, and managing patients with mild ZSD.
Article
Zellweger Syndrome (ZS) is a genetic mutation disorders with associated craniofacial and developmental anomalies in new-born babies. It also manifest with hearing and vision disorders. This case report discuss on a 2 year old male child diagnosed as ZS with hypotonia and the important milestones in the audiological diagnostic evaluation.
Article
Full-text available
Zellweger spectrum disorder (ZSD) is a rare, debilitating genetic disorder of peroxisome biogenesis that affects multiple organ systems and presents with broad clinical heterogeneity. Although severe, intermediate, and mild forms of ZSD have been described, these designations are often arbitrary, presenting difficulty in understanding individual prognosis and treatment effectiveness. The purpose of this study is to conduct a scoping review and meta-analysis of existing literature and a medical chart review to determine if characterization of clinical findings can predict severity in ZSD. Our PubMed search for articles describing severity, clinical findings, and survival in ZSD resulted in 107 studies (representing 307 patients) that were included in the review and meta-analysis. We also collected and analyzed these same parameters from medical records of 136 ZSD individuals from our natural history study. Common clinical findings that were significantly different across severity categories included seizures, hypotonia, reduced mobility, feeding difficulties, renal cysts, adrenal insufficiency, hearing and vision loss, and a shortened lifespan. Our primary data analysis also revealed significant differences across severity categories in failure to thrive, gastroesophageal reflux, bone fractures, global developmental delay, verbal communication difficulties, and cardiac abnormalities. Univariable multinomial logistic modeling analysis of clinical findings and very long chain fatty acid (VLCFA) hexacosanoic acid (C26:0) levels showed that the number of clinical findings present among seizures, abnormal EEG, renal cysts, and cardiac abnormalities, as well as plasma C26:0 fatty acid levels could differentiate severity categories. We report the largest characterization of clinical findings in relation to overall disease severity in ZSD. This information will be useful in determining appropriate outcomes for specific subjects in clinical trials for ZSD.
Article
The management of paediatric patients with inborn errors of metabolism (IEM) presents an unparalleled challenge for paediatric dentists owing to the multiplex of interrelated dental manifestations and metabolic management necessitating modifications to dental care. Inborn errors of metabolism describe a largely heterogenous group of genetic disorders namely attributable to a single gene defect essential for a specific metabolic pathway. Approximately 400 disorders have been described with an overall incidence of 1 in 5000 live births worldwide. Clinical presentation is classically inconspicuous and insidious in the neonatal period with pathophysiology attributable to accumulation of toxic by‐products which interfere with normal function, or insufficient synthesis of essential compounds. This paper aims to discuss the primary oral and maxillofacial manifestations across the scope of inborn errors of metabolism, whilst also considering how metabolic treatment has the propensity to complicate dental management. This article is protected by copyright. All rights reserved.
Article
Full-text available
Zellweger spectrum disorders (ZSD) constitute a group of rare autosomal recessive disorders characterized by a defect in peroxisome biogenesis due to mutations in one of 13 PEX genes. The broad clinical heterogeneity especially in late-onset presenting patients and a mild phenotype complicates and delays the diagnostic process. Here, we report a case of mild ZSD, due to novel PEX1 variants. The patient presented with an early hearing loss, bilateral cataracts, and leukodystrophy on magnetic resonance (MR) images. Normal results of serum very-long-chain fatty acids (VLCFA) and phytanic acid were found. Molecular diagnostics were performed to uncover the etiology of the clinical phenotype. Using whole exome sequencing, there have been found two variants in the PEX1 gene—c.3450T>A (p.Cys1150*) and c.1769T>C (p.Leu590Pro). VLCFA measurement in skin fibroblasts and C26:0-lysoPC in dried blood spot therefore was performed. Both results were in line with the diagnosis of ZSD. To conclude, normal results of routine serum VLCFA and branched-chain fatty acid measurement do not exclude mild forms of ZSD. The investigation of C26:0-lysoPC should be included in the diagnostic work-up in patients with cataract, hearing loss, and leukodystrophy on MR images suspected to suffer from ZSD.
Article
Full-text available
We performed a clinical and genetic characterization of a pediatric cohort of patients with inherited retinal dystrophy (IRD) to identify the most suitable cases for gene therapy. The cohort comprised 43 patients, aged between 2 and 18 years, with severe isolated IRD at the time of presentation. The ophthalmological characterization also included assessment of the photoreceptor layer integrity in the macular region (ellipsoid zone (EZ) band). In parallel, we carried out a targeted, next-generation sequencing (NGS)-based analysis using a panel that covers over 150 genes with either an established or a candidate role in IRD pathogenesis. Based on the ophthalmological assessment, the cohort was composed of 24 Leber congenital amaurosis, 14 early onset retinitis pigmentosa, and 5 achromatopsia patients. We identified causative mutations in 58.1% of the cases. We also found novel genotype-phenotype correlations in patients harboring mutations in the CEP290 and CNGB3 genes. The EZ band was detectable in 40% of the analyzed cases, also in patients with genotypes usually associated with severe clinical manifestations. This study provides the first detailed clinical-genetic assessment of severe IRDs with infantile onset and lays the foundation of a standardized protocol for the selection of patients that are more likely to benefit from gene replacement therapeutic approaches.
Article
Full-text available
Zellweger syndrome (ZS) is a consequence of a peroxisome biogenesis disorder (PBD) caused by the presence of a pathogenic mutation in one of the 13 genes from the PEX family. ZS is a severe multisystem condition characterized by neonatal appearance of symptoms and a shorter life. Here, we report a case of ZS with a mild phenotype, due to a novel PEX6 gene mutation. The patient presented subtle craniofacial dysmorphic features and slightly slower psychomotor development. At the age of 2 years, he was diagnosed with adrenal insufficiency, hypoacusis, and general deterioration. Magnetic resonance imaging showed a symmetrical hyperintense signal in the frontal and parietal white matter. Biochemical tests showed elevated liver transaminases, elevated serum very long chain fatty acids, and phytanic acid. After the death of the child at the age of 6 years, molecular diagnostics were continued in order to provide genetic counseling for his parents. Next generation sequencing (NGS) analysis with the TruSight One™ Sequencing Panel revealed a novel homozygous PEX6 p.Ala94Pro mutation. In silico prediction of variant severity suggested its possible benign effect. To conclude, in the milder phenotypes, adrenal insufficiency, hypoacusis, and leukodystrophy together seem to be pathognomonic for ZS.
Article
Full-text available
Peroxisomal biogenesis disorders (PBD) are caused by mutations in PEX genes, and are typically diagnosed with biochemical testing in plasma followed by confirmatory testing. Here we report the unusual diagnostic path of a child homozygous for PEX1 p.G843D. The patient presented with sensorineural hearing loss, pigmentary retinopathy, and normal intellect. After testing for Usher syndrome was negative, he was found to have PBD through a research sequencing panel. When evaluating a patient with hearing loss and pigmentary retinopathy, mild PBD should be on the differential regardless of cognitive function.
Article
Full-text available
Heimler syndrome (HS) consists of recessively inherited sensorineural hearing loss, amelogenesis imperfecta (AI) and nail abnormalities, with or without visual defects. Recently HS was shown to result from hypomorphic mutations in PEX1 or PEX6, both previously implicated in Zellweger Syndrome Spectrum Disorders (ZSSD). ZSSD are a group of conditions consisting of craniofacial and neurological abnormalities, sensory defects and multi-organ dysfunction. The finding of HS-causing mutations in PEX1 and PEX6 shows that HS represents the mild end of the ZSSD spectrum, though these conditions were previously thought to be distinct nosological entities. Here, we present six further HS families, five with PEX6 variants and one with PEX1 variants, and show the patterns of Pex1, Pex14 and Pex6 immunoreactivity in the mouse retina. While Ratbi et al. found more HS-causing mutations in PEX1 than in PEX6, as is the case for ZSSD, in this cohort PEX6 variants predominate, suggesting both genes play a significant role in HS. The PEX6 variant c.1802G>A, p.(R601Q), reported previously in compound heterozygous state in one HS and three ZSSD cases, was found in compound heterozygous state in three HS families. Haplotype analysis suggests a common founder variant. All families segregated at least one missense variant, consistent with the hypothesis that HS results from genotypes including milder hypomorphic alleles. The clinical overlap of HS with the more common Usher syndrome and lack of peroxisomal abnormalities on plasma screening suggest that HS may be under-diagnosed. Recognition of AI is key to the accurate diagnosis of HS.European Journal of Human Genetics advance online publication, 15 June 2016; doi:10.1038/ejhg.2016.62.
Article
Full-text available
Peroxisome biogenesis disorders in the Zellweger spectrum (PBD-ZSD) are a heterogeneous group of genetic disorders caused by mutations in PEX genes responsible for normal peroxisome assembly and functions. As a result of impaired peroxisomal activities, individuals with PBD-ZSD can manifest a complex spectrum of clinical phenotypes that typically result in shortened life spans. The extreme variability in disease manifestation ranging from onset of profound neurologic symptoms in newborns to progressive degenerative disease in adults presents practical challenges in disease diagnosis and medical management. Recent advances in biochemical methods for newborn screening and genetic testing have provided unprecedented opportunities for identifying patients at the earliest possible time and defining the molecular bases for their diseases. Here, we provide an overview of current clinical approaches for the diagnosis of PBD-ZSD and provide broad guidelines for the treatment of disease in its wide variety of forms. Although we anticipate future progress in the development of more effective targeted interventions, the current guidelines are meant to provide a starting point for the management of these complex conditions in the context of personalized health care.
Article
Full-text available
Zellweger spectrum disorders (ZSDs) represent the major subgroup within the peroxisomal biogenesis disorders caused by defects in PEX genes. The Zellweger spectrum is a clinical and biochemical continuum which can roughly be divided into three clinical phenotypes. Patients can present in the neonatal period with severe symptoms or later in life during adolescence or adulthood with only minor features. A defect of functional peroxisomes results in several metabolic abnormalities, which in most cases can be detected in blood and urine. There is currently no curative therapy, but supportive care is available. This review focuses on the management of patients with a ZSD and provides recommendations for supportive therapeutic options for all those involved in the care for ZSD patients.
Article
Full-text available
Heimler syndrome (HS) is a rare recessive disorder characterized by sensorineural hearing loss (SNHL), amelogenesis imperfecta, nail abnormalities, and occasional or late-onset retinal pigmentation. We ascertained eight families affected by HS and, by using a whole-exome sequencing approach, identified biallelic mutations in PEX1 or PEX6 in six of them. Loss-of-function mutations in both genes are known causes of a spectrum of autosomal-recessive peroxisome-biogenesis disorders (PBDs), including Zellweger syndrome. PBDs are characterized by leukodystrophy, hypotonia, SNHL, retinopathy, and skeletal, craniofacial, and liver abnormalities. We demonstrate that each HS-affected family has at least one hypomorphic allele that results in extremely mild peroxisomal dysfunction. Although individuals with HS share some subtle clinical features found in PBDs, the diagnosis was not suggested by routine blood and skin fibroblast analyses used to detect PBDs. In conclusion, our findings define HS as a mild PBD, expanding the pleiotropy of mutations in PEX1 and PEX6.
Article
Heimler syndrome (HS) is a rare recessive disorder characterized by sensorineural hearing loss (SNHL), amelogenesis imperfecta, nail abnormalities, and occasional or late-onset retinal pigmentation. It is the mildest form known to date of peroxisome biogenesis disorder caused by hypomorphic mutations of PEX1 and PEX6 genes. We report on a second Moroccan family with Heimler syndrome with early onset, severe visual impairment and important phenotypic overlap with Usher syndrome. The patient carried a novel homozygous missense variant c.3140T > C (p.Leu1047Pro) of PEX1 gene. As standard biochemical screening of blood for evidence of a peroxisomal disorder did not provide a diagnosis in the individuals with HS, patients with SNHL and retinal pigmentation should have mutation analysis of PEX1 and PEX6 genes.
Article
Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare co-occurence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies. This article is protected by copyright. All rights reserved.