You are on page 1of 10

Epilepsia, **(*):1–10, 2009

doi: 10.1111/j.1528-1167.2009.02066.x

FULL-LENGTH ORIGINAL RESEARCH

Partial epilepsy syndrome in a Gypsy family linked to


5q31.3-q32
*1Dora Angelicheva, yz1Ivailo Tournev, yVelina Guergueltcheva, yVioleta Mihaylova,
*Dimitar N. Azmanov, *Bharti Morar, yMelania Radionova, xShelagh J. Smith,
{Dora Zlatareva, xJohn M. Stevens, #Radka Kaneva, yVeneta Bojinova, **Kim Carter,
yyMatthew Brown, zzAssen Jablensky, *1Luba Kalaydjieva, and
x,xx1Josemir W. Sander
*Laboratory for Molecular Genetics, Centre for Medical Research and Western Australian Institute for Medical
Research, The University of Western Australia, Perth, Australia; yDepartment of Neurology, Medical University,
Sofia, Bulgaria; zDepartment of Cognitive Science and Psychology, New Bulgarian University, Sofia, Bulgaria;
xDepartment of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, United Kingdom;
{Department of Radiology, Tokuda Hospital, Sofia, Bulgaria; #Molecular Medicine Centre, Medical University,
Sofia, Bulgaria; **Western Australian Cardiovascular Genetics Group, Telethon Institute for Child Health
Research, Perth, Australia; yyCentre for Immunology and Cancer Research, Princess Alexandra Hospital,
The University of Queensland, Brisbane, Australia; zzSchool of Psychiatry and Clinical Neurosciences,
The University of Western Australia, Perth, Australia; and xxSEIN – Epilepsy Institute of The Netherlands
Foundation, Heemstede, The Netherlands

Results: We observed an early-onset partial epi-


SUMMARY
lepsy syndrome with seizure semiology strongly
Purpose: The restricted genetic diversity and suggestive of temporal lobe epilepsy (TLE), with
homogeneous molecular basis of Mendelian disor- mild intellectual deficit co-occurring in a large
ders in isolated founder populations have rarely proportion of the patients. Psychiatric morbidity
been explored in epilepsy research. Our long-term was common in the extended pedigree but did
goal is to explore the genetic basis of epilepsies in not cosegregate with epilepsy. Linkage analysis
one such population, the Gypsies. The aim of this definitively excluded previously reported loci, and
report is the clinical and genetic characterization identified a novel locus on 5q31.3-q32 with an loga-
of a Gypsy family with a partial epilepsy syndrome. rithm of the odds (LOD) score of 3 corresponding
Methods: Clinical information was collected using to the expected maximum in this family.
semistructured interviews with affected subjects Discussion: The syndrome can be classified as
and informants. At least one interictal electroen- familial temporal lobe epilepsy (FTLE) or possibly
cephalography (EEG) recording was performed a new syndrome with mild intellectual deficit. The
for each patient and previous data obtained from linked 5q region does not contain any ion channel–
records. Neuroimaging included structural mag- encoding genes and is thus likely to contribute new
netic resonance imaging (MRI). Linkage and haplo- knowledge about epilepsy pathogenesis. Identifi-
type analysis was performed using the Illumina IVb cation of the mutation in this family and in addi-
Linkage Panel, supplemented with highly informa- tional patients will define the full phenotypic
tive microsatellites in linked regions and Affyme- spectrum.
trix SNP 5.0 array data. KEY WORDS: Founder populations, Partial epi-
lepsy, Linkage analysis.

Accepted January 6, 2009; Early View publication Xxxxx xx, 200x


Address correspondence to Prof. Ley Sander, Box 29, Clinical Neuro- Epilepsy is a common neurologic disorder, with preva-
sciences Centre, UCL Institute of Neurology, Queen Square, London
WC1N 3BG, United Kingdom. E-mail: lsander@ion.ucl.ac.uk lence in some resource-poor settings of up to 1% (Sander,
1
These authors have contributed equally to the work. 2003; Duncan et al., 2006). Although exposure to pre-,
Wiley Periodicals, Inc. peri- and postnatal risk factors is likely to contribute to an
ª 2009 International League Against Epilepsy increased prevalence in some communities, there is

1
2
D. Angelicheva et al.

growing interest in the role of specific genetic factors. Phenotype characterization relied on hospital admis-
Current understanding of the molecular basis of epilepsies sions of key cases from each family, and extensive field-
is mostly confined to studies of rare large pedigrees with work involving face-to-face interviews with patients and
Mendelian forms of the disorder and mutations of large at least one key informant for each affected individual.
effect (recently reviewed by Helbig et al., 2008). Information on unaffected family members was also
Isolated founder populations have contributed to the collected from key informants. Repeat interviews were
understanding of the molecular basis of hereditary disor- conducted to check for consistency and to update informa-
ders (Peltonen et al., 1999; Ostrer, 2001; Arcos-Burgos & tion. Data on seizure history, developmental history, risk
Muenke, 2002), especially of autosomal recessive condi- factors, and medical and psychiatric comorbidity were
tions; their potential has, however, rarely been explored in collected using a semistructured interview. Affected indi-
genetic studies of epilepsy. Mendelian forms of epilepsy viduals were screened for current and past psychopathol-
are mostly autosomal dominant; however, the lack of ogy (anxiety and panic disorders, depression, mania,
effect on reproductive fitness makes it likely that founder psychotic symptoms, and disorders of memory and senso-
mutations do exist and are shared between ostensibly rium). Cognitive ability was assessed using a modified
‘‘unrelated’’ affected families, as documented for other version of the Mini Mental State Examination (MMSE),
autosomal dominant disorders (Risch et al., 1995; John adapted for population groups with high rates of social
et al., 2007). We are investigating the genetics of epilepsy and educational deprivation. All data were evaluated by
in the Bulgarian Roma/Gypsies, whose genetic structure, consensus between the two senior clinical investigators.
single-gene disorders, and founder mutations we have At least one awake interictal electroencephalography
studied for more than a decade (Kalaydjieva et al., 2001, (EEG) recording with hyperventilation was obtained for
2005). The Gypsies are a young isolate of Asian ancestry each affected individual, and previous findings were
with a history of demographic bottlenecks, strong founder retrieved from medical records. Digital EEGs were
effects, and a preserved tradition of large extended fami- acquired in accordance with international standards on the
lies, all of which facilitates genetic research (Kalaydjieva PL-EEG Wavepoint Medtronic (Minneapolis, MN,
et al., 2005). Our previous findings of restricted diversity U.S.A.) system with 25 electrode inputs (21 scalp
led us to hypothesize that the genetic basis of epilepsy may electrodes, system reference, ground, A1 and A2), and
be less heterogeneous than in outbred populations, with a analyzed independently by two neurophysiologists.
limited spectrum of founder mutations accounting for a Brain magnetic resonance imaging (MRI) was per-
large proportion of affected multiplex families and possi- formed using a 1.5T MR imager (MR Signa, GE Health-
bly also for seemingly unrelated ‘‘sporadic’’ cases, usually care, Milwaukee, WI, U.S.A.). The examination protocol
classified as genetically complex forms. Here we report consisted of five series: sagittal T1-weighted fluid-attenu-
the first results of this study—a focal epilepsy syndrome in ated inversion recovery (FLAIR); coronal T1-weighted
a Gypsy family, linked to a novel locus on chromosome 5q. three-dimensional (3D) fast spoiled gradient echo
(FSPGR), T2 FLAIR, and T2-weighted fast relaxation fast
spin echo sequence (FRFSE) and axial T2-weighted
Subjects and Methods FRFSE. The images were independently reviewed by two
General study design radiologists.
Case ascertainment relied on hospital admissions to
neurology departments (adult and pediatric) of the Medi- The family reported in this study
cal University in Sofia, general practitioners working in The S1 family (Fig. 1) is the first family enrolled in
Gypsy neighborhoods, and the network of health media- the study and has been followed since 2004. A total of
tors—members of Gypsy communities trained in primary 21 subjects from two generations participated, of whom
health care. Probands with a documented or suspected 10 (seven males and three females, aged 10–40 years)
family history of epilepsy were recruited to the study and were affected. Several additional members of this
other family members were approached. Written informed extended family (unaffected by epilepsy), for example,
consent was obtained from all adult subjects and from 2–2, 2–3, and so on (Fig. S1), declined participation in
guardians of minors. The project has been approved by, the genetic study. In the previous generation, the father
and complies with, the guidelines of the Ethics Commit- (deceased) was reported by several informants to have
tees of the Medical University in Sofia, the University of been affected. Other relatives, in more distantly related
Western Australia, and University College London. branches along the paternal line, were also described as
Data on group (subisolate) identity of affected families affected (including one who died in status epilepticus)
were collected through interviews with key informants. but were not available for investigation. No consanguin-
Family trees were based on multiple interviews, with eous marriages were reported. The pedigree structure
particular attention to consanguinity, possible multilineal indicated autosomal dominant inheritance with incom-
inheritance, and relatedness between affected families. plete penetrance.
Epilepsia, **(*):1–10, 2009
doi: 10.1111/j.1528-1167.2009.02066.x
3
Partial Epilepsy in a Gypsy Family

Figure 1.
Pedigree structure of the S1 family with 5q31.3–q32 haplotypes. The drawing includes only individuals participating in
the genetic study. Affected subjects are represented with shaded symbols, and the legend defines the shading code.
For deidentification purposes, the numbering does not follow birth order. The haplotypes in the figure include
selected microsatellites and single nucleotide polymorphisms (SNPs), illustrating the shared region and the key
recombinations.
Epilepsia ILAE

Genetic analyses Ltd., San Francisco, CA, U.S.A.). Further fine mapping of
A genome-wide scan was performed with the Illumina the 5q region was done with 23 SNPs, genotyped with the
Linkage Panel IVb, comprising 6,008 single nucleotide Affymetrix Genome-Wide SNP5.0 GeneChip array in
polymorphisms (SNPs), 5,663 of which are autosomal. samples representing the conserved and the recombinant
Genotyping data quality was checked using the Illumina haplotypes.
BeadStudio software (Illumina Inc., San Diego, CA, Linkage analysis was performed using Merlin v1.0
U.S.A.), with additional visual inspection of markers with (Abecasis et al., 2002) under a dominant model with
GenTrain or ClusterSeparation scores £0.75. Mendelian incomplete (0.7) penetrance, gene frequency 0.0001, and
inconsistencies and improbable recombinations were marker allele frequencies calculated with the Mendel
checked using the error detection function of Merlin v1.0 program (Lange et al., 2001) by maximum likelihood esti-
(Abecasis et al., 2002), and markers showing genotyping mates from the genotyping data. The calculations were
errors were excluded from subsequent analyses. done with phenocopy rates varying between 0.001 and
For the fine mapping of linked regions, we genotyped 0.01. The genetic map was as provided by Illumina
18 highly informative microsatellite markers (Table S1). (11186808_Linkage_IV_B). In the linkage analysis,
Polymerase chain reactions (PCRs) were performed under family members were defined as affected based on pres-
standard conditions, with length separation of products on ence of seizures. Unaffected subjects, including spouses,
an ABI377 DNA Analyzer or a Hitachi FMBI0II fluores- were considered unknown. The expected maximum two-
cent scanner (Hitachi Software Engineering America, point LOD score in the family, calculated for the same

Epilepsia, **(*):1–10, 2009


doi: 10.1111/j.1528-1167.2009.02066.x
4
D. Angelicheva et al.

model parameters through general simulation program for moderate intellectual deficit. Psychiatric comorbidity was
linkage analysis (SLINK) simulations (Ott, 1989; Weeks present in five subjects: bipolar affective disorder (1),
et al., 1990) was 2.96 for multiallelic markers (lower for panic disorder and mild recurrent depression (1), alcohol
the less informative SNPs). Haplotypes were constructed dependence with subdelirious psychotic episodes and
using the Merlin haplotyping function and visualized with organic personality change (1), and attention deficit
HaploPainter (Thiele & Nurnberg, 2005). The relative hyperactivity disorder (ADHD) (2). Alcohol or drug
physical positions of markers from the three datasets abuse, anxiety and depression, and ADHD were also pres-
(Illumina, microsatellites and Affymetrix) were verified ent in several unaffected members of the pedigree
in the reference sequence (NCBI NT_029289), and (Fig. S1) and did not show a pattern of cosegregation with
genetic positions were obtained from public databases or epilepsy.
determined by interpolation (for new short tandem repeat
[STR] markers). Genetic analyses
Sequencing of candidate genes was performed in three The average genotype call rate for the genome scan was
affected subjects (2–8, 3–22, and 3–23), an unaffected 0.992. Forty-six markers (0.8% of the total number of
spouse (2–19), and two normal controls from the same autosomal markers) were removed from further analysis,
population, using BigDye Terminator v3.1 (Applied based on call rates <0.9 and/or problematic clustering (21
Biosystems, Foster City, CA, U.S.A.). Exons, flanking markers) and Mendelian inconsistencies and/or double
introns, and untranslated regions of brain-specific trans- recombinations (25 markers).
cripts were PCR-amplified and sequenced with flanking Linkage analysis excluded all previously reported epi-
and internal primers (Table S1). Sequencing reactions lepsy loci (Fig. 3 and Table S2) and identified two positive
were resolved on an ABI377 DNA Analyzer and examined regions, on chromosomes 5q31.3-q32 and 10p12 (Fig. 3).
using the BioEdit software (http://www.mbio.ncsu.edu/ In the 5q region, spanning 10.6 cM (7.88 Mb), defined by
BioEdit/bioedit.html). rs2053055 on the centromeric and rs1016256 on the telo-
meric side, the highest LOD score was 3.003 at rs33388,
located at 144.52 cM. This corresponds to the maximum
Results LOD score expected in this family under the genetic
Phenotype description model and family structure described. Variations in the
The clinical data on the 10 affected individuals are sum- phenocopy rate from 0.1% to 1% did not change signifi-
marized in Table 1, with detailed descriptions available as cantly the LOD score (difference of 0.05). Independent
Supporting Information on-line. The age at onset of con- linkage analysis of the microsatellite markers, genotyped
vulsions was in childhood (1–12 years), consistently in the region, produced LOD scores of 3.00–3.01 for all
between 1 and 3 years in the third generation. In all cases, markers in the 141.8–148.82 cM interval between
the first seizure was described as convulsive, although 5q31_28TG and D5S638 (map positions specified in
other seizure types may have gone unrecognized. Uncer- Table S1).
tain data on febrile illness during initial convulsions were The combined SNP and microsatellite haplotype, co-
obtained in three patients. Consistent information on com- segregating with the disease, was inherited by all affected
plex partial seizures, commonly characterized by oroali- individuals in family S1, as well as by individual 3–17,
mentary and motor automatisms, was obtained in eight who had mental retardation but no seizures. It was not
affected individuals. Secondary generalization was found in subject 2–4, diagnosed as having organic brain
observed in all 10 individuals (description uncertain in disorder (Supporting Information). Haplotype examina-
one), with a frequency of 1–3 per month. Aura was auto- tion identified a key centromeric recombination in indi-
nomic in five cases, epigastric in four, and somatosensory vidual 3–25 and a telomeric recombination in individual
in four; cephalic, dyscognitive, illusory, and auditory 3–27 (Figs. 1 and 4). Fine mapping placed the centromeric
warning signs were also described. Interictal EEGs were recombination in the interval between 5q31_18CA and
abnormal in five patients, with localization mostly sug- 5q31_28TG, and the telomeric between rs728937 and
gesting temporal lobe involvement, and abnormalities also D5S434, reducing the size of the critical gene region to
recorded in the centroparietal and frontal regions (Fig. 2, 8.47 cM or 5.7 Mb. Further breakpoint mapping, using
Table 1 and Supporting Information). Antiepileptic drugs the Affymetrix SNP5.0 array, shifted the telomeric bound-
had been prescribed, but it is probable that adherence to ary proximal to rs7709303 and decreased slightly the size
treatment had been poor and some patients had never been of the interval to 7.80 cM or 5.6 Mb (Fig. 4), but did not
treated. Neuroimaging revealed no structural lesions or contribute new information for the centromeric recombi-
asymmetries. Developmental delay and/or learning diffi- nation.
culties (commonly attributed to memory problems and A second, minor linkage peak was observed on chromo-
school drop-out) were reported in seven patients, of whom some 10p12, spanning 6.34 cM (3.9 Mb) between
four (subjects 2–8, 3–18, 3–23, and 3–26) had mild to rs1928365 and rs913034 (both flanked by uninformative
Epilepsia, **(*):1–10, 2009
doi: 10.1111/j.1528-1167.2009.02066.x
Table 1. Clinical characteristics of the partial epilepsy syndrome in family S1
Year Seizure Simple
Case of onset partial Complex partial Brain
ID birth Sex (age) FS seizures seizures Aura features Generalization Semiology EEG imaging Comorbidity
2–6 1968 F 7 years None None None reported Epigastric 1–2/month (daytime) T 2006 & 2007 normal MRI normal Hypertension,
reported reported obesity, panic
attacks
2–8 1972 M 2 years None None Ambulatory automatisms Somatosensory, 2/month (daytime) T/E-T 2007 normal MRI normal Alcohol abuse,
reported reported (runs out into the street, autonomic, Mixed subdelirious
urinates; disoriented) dyscognitive partial episodes; organic
personality
change; mild ID
2–10 1975 F 12 None None Ambulatory (ran out into Somatosensory, 1–3/month (daytime, T/E-T 2001 F-T sharp-slow CT normal Bipolar disorder;
years reported reported the street naked); epigastric, possibly also Mixed wave discharges; obsessive-
oroalimentary auditory nocturnal) partial generalization 2006 compulsive
automatisms R&L F sharp waves features
3–16 1989 M 3 years None None Manual automatisms (pulls Autonomic, 1/month (daytime) T 2004 R P-T spike-slow CT&MRI None reported
reported reported down curtains, breaks epigastric, waves 2006 L F-T normal
glasses, grabs things); dyscognitive theta; R & L F spikes
dysphasic automatisms
(talks incomprehensively)
3–18 1988 M 2 years Possible None Ambulatory automatisms None reported Uncertain T 2006 normal MRI normal Moderate ID,
reported (runs around, waving congenital limb
his hands), disoriented malformation
3–22 1995 F 1½ Possible Cephalic, Orofacial and mimetic Somatosensory 2–3/month (both T 2006 R&L C-P sharp CT & MRI Delayed
year epigastric, automatisms (dizziness), daytime and waves 2007 R T normal developmental
illusory oroalimentary nocturnal) sharp–slow waves milestones;
stammer
3–23 1997 M 1 year Possible None Uncertain Aura present, < 1/month (daytime T 2006 R C-T focal CT & MRI Delayed
reported description and nocturnal) abnormalities; 2006 normal developmental
uncertain & 2007 normal milestones.
Mild ID
3–25 1998 M 1 year None None Manual and ambulatory Autonomic 1/week E-T 2006 Bilateral CT & MRI ADHD (constant
reported reported automatisms (running and polyspike waves; R F normal agitation,
screaming, pulling objects) focus 2007 normal aggressive
behaviour)
3–26 1995 M 1 year None None Ambulatory, (‘‘running Autonomic, 1/week T 2005, 2006, 2007 MRI normal Mild ID
reported reported around like a horse’’) and epigastric, normal
vocal (screaming) mimetic
automatisms
3–27 1992 M Infancy None None Dyscognitive (incoherent Somatosensory 1–2/month (both Mixed 2004, 2006, 2007 CT & MRI Learning
reported reported speech); interactive cephalic, daytime and partial normal normal (memory)
automatisms (talks to self, illusory, nocturnal) difficulties
grimacing) autonomic
Partial Epilepsy in a Gypsy Family

T, temporal; E-T, extratemporal; R, right; L, left; C, central; F, frontal; P, parietal; ID, intellectual deficit; FS, febrile seizures.

doi: 10.1111/j.1528-1167.2009.02066.x
Epilepsia, **(*):1–10, 2009
5
6
D. Angelicheva et al.

Figure 2.
(A) Case 3–25. Age 8 at time of interictal electroencephalography (EEG) recording. The EEG shows an isolated burst
of generalized spike-wave discharge. There were no focal findings in this trace. (B) Case 3–22. Age 13 years.
Interictal recording showing biparietal focal abnormalities: paroxysmal slow complex on the left (P3), and low
amplitude sharp transients on the right (P4).
Epilepsia ILAE

Epilepsia, **(*):1–10, 2009


doi: 10.1111/j.1528-1167.2009.02066.x
7
Partial Epilepsy in a Gypsy Family

Figure 3.
Genome-wide multipoint parametric linkage analysis results. Individual chromosomes are separated with dashed
vertical lines. Genotyping was done using the Illumina Linkage Panel IVb. Linkage analysis was performed with Merlin
v1.0.
Epilepsia ILAE

Figure 4.
The region on 5q31.3–q32, showing linkage to the partial epilepsy syndrome in family S1. Markers are presented
following their physical map order from centromere to telomere. Single nucleotide polymorphism (SNP) alleles
correspond to the variant on the positive strand, as shown in dbSNP (http://www.ncbi.nlm.nih.gov/projects/SNP/).
Microsatellite alleles are numbered in ascending size order. The key recombinations in subjects 3–25 and 3–27 are
shown as filled bars. The 13 brain-expressed genes, contained in the linked region, are shown at the bottom of the
figure.
Epilepsia ILAE

Epilepsia, **(*):1–10, 2009


doi: 10.1111/j.1528-1167.2009.02066.x
8
D. Angelicheva et al.

markers). The highest LOD score was 0.94, at rs997462 several years in the reports of different informants.
(51.93 cM). Genotyping and independent linkage analysis Another feature that requires cautious interpretation is
of more informative microsatellite markers resulted in intellectual disability, where an adverse social environ-
negative LOD scores ()1.7 to )3.8), excluding 10p12 as a ment and lack of culturally fair psychological tests may
potential modifying locus. significantly affect diagnostic assessment. We are aware
According to the current build 36.3 of the human gen- of the risk of such bias and have conservatively
ome, the 5.6 Mb interval on 5q31.3–32 contains 31 constrained our interpretation of those aspects of the
validated and predicted genes, of which 13 are expressed phenotype to verifiable facts. The data on the remaining
in brain (Fig. 4). Three were ranked as the best functional key aspects of the clinical phenotypes, however, were
candidates: Potassium Channel Tetramerization Domain usually consistent and reliably reproduced in several
16 (KCTD16), Fibroblast Growth Factor (FGF1) and interviews.
Sprouty homologue 4 (SPRY4). Sequencing analysis of To summarize, we have identified a Gypsy family with
the coding and untranslated regions of KCTD16 identified an epilepsy syndrome of autosomal dominant inheritance
10 known polymorphisms listed in the public databases and reduced penetrance, with onset in the first decade of
(rs3797084, rs187034, rs349704, rs349703, rs358651, life, complex partial seizures, and frequent secondary
rs349701, rs349700, rs349699, rs349698, rs349697) and generalization. The complex partial seizures had mostly
three novel sequence variants—a synonymous coding temporal lobe characteristics, but in some cases extra-
SNP (S303S; c.909C>T) and two changes in the 3¢UTR temporal phenomenology was also observed. Interictal
(c. 1287 + 15C>T and c. 1287 + 3273 G>A). Lack of co- EEG abnormalities were present in half of the patients
segregation with the disease phenotype and presence (Fig. 2), and at some stage localized in the temporal or
in the unaffected controls ruled out their role as disease- adjacent areas. Discharges with frontal localization or a
causing mutations. frontal maximum were observed in three patients, two of
whom (2–10 and 3–16) had frontotemporal and parieto-
temporal abnormalities in previous EEGs and seizure
Discussion semiology most characteristic of temporal lobe epilepsy
Epilepsy is among the best examples of extensive (Table 1 and Supporting Information). Brain imaging
genetic and clinical heterogeneity, with multiple loci, showed no structural abnormalities or asymmetries.
incomplete penetrance, low-grade genotype–phenotype Intellectual disability in the mild to moderate range
correlations, and a limited number of large informative meeting ICD-10 criteria was established in 4 of 10
pedigrees, all contributing to the difficulty in elucidating subjects with epilepsy, and, with certainty, in only one of
its molecular basis. Founder populations could facilitate the unaffected individuals, thus suggesting a possible
research efforts if their restricted diversity and shared cosegregation with the epileptic syndrome. Other psychi-
ancestral mutations also apply to epilepsy. Our project atric and behavioral morbidity in this family, although
aims to test this possibility in the Gypsy population, where common, did not appear to cosegregate with seizure
our previous studies have shown a strong founder effect disorders (Fig. S1).
and allowed assessment of the spectrum of clinical mani- This condition does not resemble partial epilepsy with
festations in genetically homogeneous groups (Tournev pericentral spikes (PEPS) (Kinton et al., 2002) and the
et al., 1999; Thomas et al., 2001; Colomer et al., 2006; differential diagnosis includes familial temporal lobe
Mihaylova et al., 2007). The well-known constraints on epilepsy (FTLE) and familial partial epilepsy with vari-
conducting genetic research into epilepsy, such as wide- able foci (FPEVF), both showing clinical as well as
spread fear of stigmatization and the difficulty in eliciting genetic heterogeneity (Berkovic et al., 1996, 2004; Cen-
‘‘crisp’’ phenotypes, were also encountered in this study. des et al., 1998; Scheffer et al., 1998; Xiong et al., 1999;
They were augmented by the social marginalization of the Gambardella et al., 2000; Picard et al., 2000; Callenbach
Gypsies, and the need for adapting the clinical investiga- et al., 2003; Claes et al., 2004). Individual members in
tions to the cultural tradition, concepts, and beliefs of this FPEVF families are described as having a distinct elec-
population, as well as educational and economic depriva- troclinical phenotype, with congruence between clinical
tion. In the course of the project, we have improved data and EEG localization. The most common seizure type in
collection by fine-tuning the interview and adapting it to some families appears to be nocturnal FLE. Here the
the cultural idiom, and by using multiple sources of infor- affected individuals do not have such distinct electro-
mation and repeat interviews on each affected subject. clinical phenotypes; some may have mixed seizure types
The quality of information remained uncertain regarding (extratemporal and temporal) and their EEG focus tends
several specific clinical parameters: a history of febrile not to be specifically congruent with seizure semiology.
convulsions has proved very difficult to verify, and the The lack of ictal EEG recordings makes it difficult to
timing of specific events, particularly age at onset of sei- establish a more definitive syndromic diagnosis.
zures, was occasionally discrepant, with a difference of Although the preceding differential diagnoses refer to
Epilepsia, **(*):1–10, 2009
doi: 10.1111/j.1528-1167.2009.02066.x
9
Partial Epilepsy in a Gypsy Family

forms of idiopathic partial epilepsy, it is possible that Australia and by UCLH/UCL, which received a proportion of funding
from the Department of Health’s NIHR Biomedical Research Centre’s
the syndrome in family S1 also includes a raised inci- funding scheme.
dence of intellectual deficit. The interpretation of our
We confirm that we have read the Journal’s position on issues involved in
findings should take into account a number of possible ethical publication and affirm that this report is consistent with those
confounders: the generally high frequency of affective guidelines.
and anxiety disorders in epilepsy patients (Kanner, 2007, Disclosure: None of the authors has any conflict of interest in regard to
2008), a social environment that could lead to dysthymic this work to disclose.
depression and dropping out of school, and widespread
alcohol abuse in this population. The identification of
the disease mutation and of additional patients carrying
References
the same mutation should contribute to a more precise Abecasis GR, Cherny SS, Cookson WO, Cardon LR. (2002) Merlin –
definition of the full phenotypic spectrum. rapid analysis of dense genetic maps using sparse gene flow trees.
Nat Genet 30:97–101.
Our genetic analysis excluded all previously reported Arcos-Burgos M, Muenke M. (2002) Genetics of population isolates.
loci, including those linked to FTLE and FPEVF (10q24, Clin Genet 61:233–247.
4q13.2-q21.3, 12q22-q23.3, 18qter and 1q25-q31, 2qter, Berkovic SF, McIntosh A, Howell RA, Mitchell A, Sheffield l, Hopper
JL. (1996) Familial temporal lobe epilepsy: a common disorder iden-
22q11-q12) with LOD scores lower than )3 (Fig. 3 and tified in twins. Ann Neurol 40:227–235.
Table S2). Berkovic SF, Serratosa JM, Phillips HA, Xiong L, Andermann E,
The results tentatively place the disease gene in a Diaz-Otero F, Gomez-Garre P, Martin M, Fernandez-Bullido Y,
Andermann F, Lopez-Cendes I, Dubeau F, Desbiens R, Scheffer I,
5.6 Mb interval on 5q31.3-q32, centromeric of a cluster of Wallace RH, Mulley JC, Pandolfo M. (2004) Familial partial epilepsy
GABA-A (c-aminobutyric acid A) receptor genes, the with variable foci: clinical features and linkage to chromosome
involvement of which was excluded by multiple recombi- 22q12. Epilepsia 45:1054–1060.
Callenbach PM, van der Maagdenberg AM, Hottenga JJ, van den
nations in the S1 family. The best candidate genes within Boogerd EH, de Coo RF, Lindhout D, Frants RR, Sandkuijl LA,
the 5q locus include KCTD16, FGF1, and SPRY4. The Brouwer OF. (2003) Familial partial epilepsy with variable foci in a
choice of Potassium Channel Tetramerization Domain 16 Dutch family: clinical characteristics and confirmation of linkage to
chromosome 22q. Epilepsia 44:1298–1305.
gene (KCTD16) as a candidate gene is based on its pattern Cendes F, Lopes-Cendes I, Andermann E, Andermann F. (1998) Familial
of brain expression in the amygdalae, caudate nucleus, temporal lobe epilepsy: a clinically heterogeneous syndrome. Neuro-
and hippocampus (Nagase et al., 2000) and on the involve- logy 50:554–557.
Claes L, Audenaert D, Deprez L, Van Paesschen W, Depondt C,
ment of another member of the same gene family, KCTD7, Goossens D, Del-Favero J, Van Broeckhoven C, De Jonghe P. (2004)
in autosomal recessive progressive myoclonic epilepsy Novel locus on chromosome 12q22–q23.3 responsible for familial
with dementia (Van Bogaert et al., 2007). Although no temporal lobe epilepsy associated with febrile seizures. J Med Genet
41:710–714.
disease-causing mutation was identified in the KCTD16 Colomer J, Gooding R, Angelicheva D, King RH, Guillen-Navarro
coding sequence, our current data do not rule out the possi- E, Parman Y, Nascimento A, Conill J, Kalaydjieva L. (2006)
bility of an intronic mutation causing aberrant splicing or Clinical spectrum of CMT4C disease in patients homozygous for
the p.Arg1109X mutation in SH3TC2. Neuromuscul Disord 16:449–
affecting a regulatory sequence. Further analysis is in pro- 453.
gress. The acidic Fibroblast Growth Factor (FGF1) has Cuevas P, Gimenez-Gallego G. (1997) Role of fibroblast growth factors
been found to suppress convulsions in kainate-induced in neural trauma. Neurol Res 19:254–256.
Duncan JS, Sander JW, Sisodiya SM, Walker MC. (2006) Seminar: adult
epilepsy in rats (Cuevas & Gimenez-Gallego, 1997), pro- epilepsy. Lancet 367:1087–1100.
mote the survival of hippocampal pyramidal neurons after Gambardella A, Messina D, Le Piane E, Oliveri RL, Annesi G, Zappia
ischaemia (Sasaki et al., 1992), and enhance long-term M, Andermann E, Quattrone A, Aguglia U. (2000) Familial temporal
lobe epilepsy autosomal dominant inheritance in a large pedigree
potentiation (Sasaki et al., 1994; Reuss & von Bohlen und from southern Italy. Epilepsy Res 38:127–132.
Halbach, 2003). The Sprouty 4 gene (SPRY4) is involved Helbig I, Scheffer IE, Mulley JC, Berkovic SF. (2008) Navigating the
in the regulation of FGF function via inhibition of the channels and beyond: unravelling the genetics of the epilepsies.
Lancet Neurol 7:231–245.
downstream ras/MAP kinase signaling pathway (Leeksma John EM, Miron A, Gong G, Phipps AI, Felberg A, Li FP, West DW,
et al., 2002). Given the lack in the 5q locus of obvious epi- Whittemore AS. (2007) Prevalence of pathogenic BRCA1 mutation
lepsy candidate genes encoding ion channels, the identifi- carriers in 5 US racial/ethnic groups. JAMA 298:2869–2876.
Kalaydjieva L, Gresham D, Calafell F. (2001) Genetic studies of the
cation of the novel gene has the potential of contributing Roma (Gypsies): a review. BMC Med Genet 2:5.
new information about the molecular pathogenesis of Kalaydjieva L, Morar B, Chaix R, Tang H. (2005) A newly discovered
epilepsy. founder population: the Roma/Gypsies. BioEssays 27:1084–1094.
Kanner AM. (2007) Epilepsy and mood disorders. Epilepsia 48(Suppl
9):20–22.
Kanner AM. (2008) Mood disorder and epilepsy: a neurobiologic per-
Acknowledgments spective of their relationship. Dialogues Clin Neurosci 10:39–45.
Kinton L, Johnson MR, Smith SJ, Farrell F, Stevens J, Rance JB,
We wish to thank the families participating in this study, the Neurode- Claudino AM, Duncan JS, Davis MB, Wood NW, Sander JW. (2002)
generative Disorders Centre, QEII Medical Centre for the Affymetrix Partial epilepsy with pericentral spikes: a new familial epilepsy syn-
SNP5.0 array genotyping, and the Western Australian Genetic Epidemi- drome with evidence for linkage to chromosome 4p15. Ann Neurol
ology Resource for bioinformatics support. The study was funded by 51:740–749.
grant 458736 of the National Health and Medical Research Council of

Epilepsia, **(*):1–10, 2009


doi: 10.1111/j.1528-1167.2009.02066.x
10
D. Angelicheva et al.

Lange K, Cantor R, Horvath S, Perola M, Sabatti C, Sinsheimer J, Sobel L, Shmarov A, Muddle JR, Nourallah M, Tournev I. (2001) Heredi-
E. (2001) Mendel version 4.0: A complete package for the exact tary motor and sensory neuropathy-Russe: new autosomal recessive
genetic analysis of discrete traits in pedigree and population data sets. neuropathy in Balkan Gypsies. Ann Neurol 50:452–457.
Am J Hum Genet 69(Suppl):A1886. Tournev I, Kalaydjieva L, Youl B, Ishpekova B, Guerguelcheva V,
Leeksma OC, Van Achterbetg TA, Tsumura Y, Toshima J, Eldering E, Kamenov O, Katzarova M, Kamenov Z, Raicheva-Ternzieva M,
Kroes WG, Mellink C, Spaargaren M, Mizuno K, Pannekoek H, de King RHM, Petkov R, Shmarov A, Dimitrova G, Popova N, Uzunova
Vries CJ. (2002) Human sprouty 4, a new ras antagonist on 5q31, M, Milanov S, Petrova J, Petkov Y, Kolarov G, Anev L, Radeva O,
interacts with the dual specificity kinase TESK1. Eur J Biochem Thomas PK. (1999) Congenital Cataracts Facial Dysmorphism
269:2546–2556. Neuropathy (CCFDN) syndrome, a novel complex genetic disease in
Mihaylova V, Hantke J, Sinigerska I, Cherninkova S, Raicheva M, Balkan Gypsies: clinical and electrophysiological observations. Ann
Bouwer S, Tincheva R, Khuyomdziev D, Bertranpetit J, Chandler D, Neurol 45:742–750.
Angelicheva D, Kremensky I, Seeman P, Tournev I, Kalaydjieva L. Van Bogaert P, Azizieh R, Desir J, Aeby A, De Meirleir L, Laes JF,
(2007) Highly variable neural involvement in sphingomyelinase- Christiaens F, Abramowicz MJ. (2007) Mutation of a potassium
deficient Niemann-Pick disease caused by an ancestral Gypsy muta- channel-related gene in progressive myoclonic epilepsy. Ann Neurol
tion. Brain 130:1050–1061. 61:579–586.
Nagase T, Kikuno R, Ishikawa KI, Hirosawa M, Ohara O. (2000) Predic- Weeks DE, Ott J, Lathrop GM. (1990) SLINK: a general simulation pro-
tion of the coding sequences of unidentified human genes XVI. The gram for linkage analysis. Am J Hum Genet 47(Suppl 3):A204.
complete sequences of 150 new cDNA clones from brain which code Xiong L, Labuda M, Li DS, Hudson TJ, Desbiens R, Patry G, Verret S,
for large proteins in vitro. DNA Res 7:65–73. Langevin P, Mercho S, Seni M-H, Scheffer I, Dubeau F, Berkovic
Ostrer H. (2001) A genetic profile of contemporary Jewish populations. SF, Andermann F, Andermann E, Pandolfo M. (1999) Mapping of a
Nat Rev Genet 2:891–898. gene determining familial partial epilepsy with variable foci to chro-
Ott J. (1989) Computer-simulation methods in human linkage analysis. mosome 22q11–q12. Am J Hum Genet 65:1698–1710.
Proc Natl Acad Sci U S A 86:4175–4178.
Peltonen L, Jalanko A, Varilo T. (1999) Molecular genetics of the Finnish
disease heritage. Hum Mol Genet 8:1913–1923. Supporting Information
Picard F, Baulac S, Kahane P, Hirsch E, Sebastianelli R, Thomas P,
Vigevano F, Genton P, Guerrini R, Gericke CA, An I, Rudolf G, Her-
mann A, Brice A, Marescaux C, LeGuern E. (2000) Dominant partial Additional supporting information may be found in the
epilepsies. A clinical, electrophysiological and genetic study of 19 online version of this article:
European families. Brain 123:1247–1262.
Reuss B, von Bohlen und Halbach O. (2003) Fibroblast growth factors Figure S1: Pedigree structure of the extended S1 family
and their receptors in the central nervous system. Cell Tissue Res (including the branches with epilepsy analyzed in this
313:139–157. study), with different symbols indicating the phenotypes
Risch N, de Leon D, Ozelius L, Kramer P, Almasy L, Singer B, Fahn S,
Breakefield X, Bressman S. (1995) Genetic analysis of idiopathic tor- of epilepsy and comorbidities: mild intellectual deficit
sion dystonia in Ashkenazi Jews and their recent descent from a small and psychiatric disorders [mood disorders, attention defi-
founder population. Nat Genet 9:152–159. cit hyperactivity disorder (ADHD), alcohol and drug
Sander JW. (2003) The epidemiology of the Epilepsies revisited. Curr
Opin Neurol 16:165–170. abuse].
Sasaki K, Oomura Y, Suzuki K, Hanai K, Yagi H. (1992) Acidic fibro-
blast growth factor prevents death of hippocampal CA1 pyramidal
Table S1: Details on primers used for fine mapping and
cells following ischemia. Neurochem Int 21:397–402. candidate gene sequencing.
Sasaki K, Oomura Y, Figurov A, Yagi H. (1994) Acidic fibroblast growth
factor facilitates generation of long-term potentiation in rat hippo- Table S2: Known epilepsy loci with LOD scores in the
campal slices. Brain Res Bull 33:505–511. investigated Gypsy family S1.
Scheffer IE, Phillips HA, O’Brien CE, Saling MM, Wrennall JA, Wallace
RH, Mulley JC, Berkovic SF. (1998) Familial partial epilepsy with Please note: Wiley-Blackwell is not responsible for the
variable foci: a new partial epilepsy syndrome with suggestion of content or functionality of any supporting information
linkage to chromosome 2. Ann Neurol 44:890–899. supplied by the authors. Any queries (other than missing
Thiele H, Nurnberg P. (2005) HaploPainter: a tool for drawing pedigrees
with complex traits. Bioinformatics 21:1730–1732. material) should be directed to the corresponding author
Thomas PK, Kalaydjieva L, Youl B, Rogers T, Angelicheva D, King RH, for the article.
Guergueltcheva V, Colomer J, Lupu C, Corches A, Popa G, Merlini

Epilepsia, **(*):1–10, 2009


doi: 10.1111/j.1528-1167.2009.02066.x

You might also like