You are on page 1of 10

Review

Progressive myoclonic epilepsies: a review of genetic and


therapeutic aspects
Amre Shahwan, Michael Farrell, Norman Delanty Lancet Neurol 2005; 4: 239–48
Department of Neurology and
The progressive myoclonic epilepsies (PMEs) are a group of symptomatic generalised epilepsies caused by rare Neuroscience (A Shahwan
MRCPCH, N Delanty FRCPI); and
disorders, most of which have a genetic component, a debilitating course, and a poor outcome. Challenges with PME Department of Pathology
arise from difficulty with diagnosis, especially in the early stages of the illness, and further problems of management (Neurology), Beaumont
and drug treatment. Recent advances in molecular genetics have helped achieve better understanding of the different Hospital, Dublin, Ireland
disorders that cause PME. We review the PMEs with emphasis on updated genetics, diagnosis, and therapeutic options. (Prof M Farrell FRCPath)
Correspondence to:
Dr Norman Delanty, Department
Progressive myoclonic epilepsies (PMEs) are and mild dementia is typically a late feature. However,
of Neurology, Beaumont
characterised by myoclonic seizures, tonic-clonic seizures, this is not constant and progress in the genetic basis of Hospital, Dublin 9, Ireland
and progressive neurological deterioration, typically with the disorder has led to finding atypical evolutions even normandelanty@eircom.net
cerebellar signs and dementia. In different disease within the same family, with some affected individuals
entities, various types of seizures and neurological displaying severe cognitive involvement. With advances
symptoms also occur, which may guide the clinician to in antiepileptic drug development and their use, the
the most likely cause. Myoclonus in PME is typically prognosis of the disease has improved significantly and
fragmentary and multifocal, and is often precipitated by patients now live into their sixties.7
posture, action, or external stimuli such as light, sound, or The EEG background may be normal during the first
touch. It is particularly apparent in musculature of the years of the disease. EEG features may mimic completely
face and distal extremities. Bilateral massive myoclonic that of idiopathic generalised epilepsy at the beginning of
jerks that tend to involve muscles of proximal limbs may the disease. With progress of the illness, it becomes
also occur.1 With the disabling myoclonus there is the risk abnormal with diffuse slow background activity and
of injury and impairment of activities of daily living. The generalised high-voltage spike and wave, and polyspike
age of onset, presenting symptoms, predominance of and wave paroxysms, ranging from a slow frequency of
symptoms as seizures, or myoclonus over cerebellar signs 2–3 Hz to faster frequencies of 4–6 Hz,8,9 which reach a
and dementia vary substantially across the different maximum anteriorly. Photosensitivity is typical.10 MRI of
disorders. There are five main causes of PME that have the brain may be normal or can show reduced bulk of the
been more accurately defined with recent advances in basis pontis, medulla, and cerebellar hemispheres, and
genetic studies (panel 1). less often, cerebral atrophy.11

Unverricht-Lundborg disease Genetics and diagnosis


Unverricht-Lundborg disease (ULD), or epilepsy ULD is linked to chromosome 21q22.3.12 Further linkage
progressive myoclonus type 1, is an autosomal-recessive disequilibrium and historical recombination breakpoint
disorder that was described by Unverricht2 in 1891, and mapping placed the associated gene, CSTB (formerly
by Lundborg3 in 1903. It is the most common PME. It
was initially recognised as a geographic cluster in
Finland, where the prevalence is one in 20 000 births.4 Panel 1: Definite diagnosis of specific types of PME
Clusters of a phenotypically and sometimes genetically
Unverricht-Lundborg disease
identical disorder occur in southern Europe and north
CSTB gene mutation
Africa,5 and sporadically worldwide.
Lafora disease
Clinical picture and investigations Lafora bodies in skin biopsy or EPM2A mutation
The age of symptom onset in ULD is 6–15 years.6
Myoclonic epilepsy with ragged red fibres
Symptoms progress insidiously. Stimulus-sensitive
Ragged red fibres in muscle biopsy or MTTK mutation
myoclonic jerks are an essential feature for the diagnosis
of the disease and are the first symptom in at least half of Neuronal ceroid lipofuscinosis
patients.7 The myoclonus is generally quite severe and Typical intracellular inclusions or mutation in TPP1, CLN3, and
shares the common characteristics of myoclonus in CLN5
other PMEs. Generalised tonic-clonic seizures are the
Sialidoses
presenting feature in many patients,8 but in rare cases
Neuraminidase deficiency in leucocytes or fibroblasts
these may not occur. Absence seizures may also be
observed. Neurological examination is initially normal, Dentatorubral-pallidoluysian atrophy
but patients later develop ataxia, incoordination, Abnormal CAG repeats
intention tremor, and dysarthria. Progression to ataxia

http://neurology.thelancet.com Vol 4 April 2005 239


Review

known as EPM1), between regions D21S2040 and Clinical picture and investigations
D21S1259.13 CSTB encodes cystatin B, a cysteine In most patients, the onset of progressively worsening
protease inhibitor.14 The main mutation in CSTB, even seizures occurs age 12–17 years. Before that, physical
among patients of different ethnic origins, is an unstable and mental development is within normal limits.26 Many
expansion of a dodecamer repeat (CCCCGCCCCGCG) patients experience isolated febrile and non-febrile
in the 5´ untranslated promoter region.15 The range of convulsions earlier in childhood.27 At onset it may be
normal alleles (repeats) is two to three copies, but difficult to distinguish Lafora’s disease clinically from
expanded alleles associated with the disease phenotype typical idiopathic generalised epilepsy with no evidence
contain at least 30 copies. Five other mutations that of cognitive decline, particularly juvenile myoclonic
affect one or two nucleotides in CSTB cause the disease epilepsy. Seizure types in Lafora’s disease include
in a few patients.16 Affected individuals may have myoclonus, occipital seizures with transient blindness
expansion of the dodecamer on both alleles, expansion and visual hallucinations, atypical absences, and atonic
on one and point mutation on the other, or, rarely, a and complex partial seizures. As the disease progresses,
point mutation on both alleles. Cystatin B inhibits the seizures become more intractable and status epilepticus
papain family of cysteine proteases, which are involved of any seizure type is not unusual.28,29 Cognitive decline,
in the initiation of apoptosis. The exact pathophysiology dysarthria, and ataxia appear early. Emotional
of the disease remains unknown. Presumably, with disturbance is common early in the disease and
mutations in cystatin B and loss of its inhibition of dementia develops gradually. Later, patients are disabled
cysteine proteases, apoptosis proceeds abnormally and with continuous myoclonus.30 Most patients die within
neurodegeneration results.17 A multiprotein complex has 10 years of onset.31 Early in Lafora’s disease, the EEG has
been identified to interact with cystatin B in vitro.18 a well-organised background with multiple spike-and-
Immunofluorescent confocal microscopy showed that wave discharges, and photosensitivity is common;
the same proteins are present in the granule cells of the however, induced epileptiform discharges may occur
developing cerebellum and the purkinje cells of adult rat only at low frequency (1–6 Hz) of photic stimulation.32
cerebellum, which raises the possibility that a cystatin B Erratic myoclonus may be seen without EEG correlation.
multiprotein complex may have a specific cerebellar Spike-and-wave discharges are not accentuated during
function.18 For diagnosis, clinical suspicion is sleep. During the next few months to years, the
paramount. Detection of the mutation in the cystatin B background deteriorates and sleep features are lost.
gene can be used to confirm the diagnosis (panel 1). Multifocal epileptiform abnormalities, mainly occipital
in location, appear in addition to generalised bursts.33 In
Treatment longitudinal EEG studies, the spike-and-wave pattern
There is no specific treatment for ULD, and palliative and changes from a frequency of 3 Hz in the early stages to
rehabilitative management is therefore the mainstay of faster frequencies of 6–12 Hz as the disease progresses.34
patient care. For seizure and myoclonus control, valproic Lafora bodies, are present in neurons, but can be seen in
acid has traditionally been the drug of choice, especially if many other tissues such as skin, liver, and muscle,
started early in the course of the disease.19 Clonazepam although diagnosis is most conveniently made by
may also help as an add-on therapy.19 High-dose examination of the eccrine ducts of the sweat glands in a
piracetam also helps in many patients.20 Levetiracetam skin biopsy specimen.35 Lafora bodies may range in size
had a substantial beneficial effect on a female patient from a barely visible 1 m dot to a large, rounded
with ULD. She was able to walk assisted and write her intracytoplasmic basophilic structure (figure 1).
name, after being wheelchair bound with severe
stimulus-evoked and action-induced multifocal Genetics and diagnosis
myoclonus.9 The antimyoclonic effect of levetiracetam in Lafora’s disease is an autosomal recessive disorder. Up
13 patients with ULD was most effective when started to 80% of patients with the disorder have a mutation in
early in the course of the disease.21 Zonisamide has been the EPM2A gene on chromosome 6q at locus 24.36 The
used with success.22 Phenytoin may significantly worsen gene encodes laforin, a dual-specificity protein tyrosine
myoclonus and precipitate cerebellar signs and cerebellar phosphatase, primarily associated with ribosomes. The
atrophy.23 Myoclonus in a patient treated with vagus- function of laforin is largely unknown but involvement
nerve stimulation also improved.24 in translational regulation and protein folding may
underlie the molecular basis of the disease.37 Laforin
Lafora’s disease may also cause glycogen synthase hyperfunction,
First described by Lafora and Gluelkin in 1911, Lafora’s contributing to the endoplasmic-reticulum-associated
disease is characterised by epilepsy, myoclonus, polyglucosan depositions seen in the disease.30 More
dementia, and Lafora bodies, which are periodic- recently, a new Lafora’s disease locus, NHLRC1
acid–Schiff-positive intracellular polyglucosan inclusion (formerly EPM2B), has been mapped to a 2·2 Mbp
bodies found in neurons, heart, skeletal muscle, liver, region at 6p22, a region that codes for several proteins,
and sweat-gland duct cells.25 including kinesins, which play a major part in axonal

240 http://neurology.thelancet.com Vol 4 April 2005


Review

and dendritic transport in neurons (table 1).38 Evidence


for a third locus for Lafora’s disease, yet to be identified,
has been recently reported.39
The narrow age of onset, progressive dementia, rapid
and relentless progression to death, and frequent
occipital seizures are clinical clues to diagnosis.
Diagnosis is confirmed by detecting Lafora bodies in
skin biopsy specimens, but this is likely to be superseded
by analysis of the EPM2A gene, yielding a known
mutation in 80% of patients with Lafora’s disease.

Treatment
Treatment for Lafora’s disease remains palliative.
Advances in gene, protein, or stem-cell therapies may
allow replacement of the defective protein in the future.
Knockout mouse and natural canine models of the
disease may yield new insights into pathogenesis, and
may facilitate the testing of new therapeutic approaches.
Meanwhile, on the basis of the knowledge that Lafora
Figure 1: Basophilic neuronal intracytoplasmic spherical Lafora bodies
bodies are carbohydrate compounds, an international
These may vary in size and degree of basophilia, and are distributed through the cortex, substantia nigra, thalamus,
collaborative clinical trial based at the US National and cerebellar cortex. Muscle, liver, and skin appendages can also contain Lafora bodies.
Institutes of Health has been begun to test whether
carbohydrate restriction will diminish Lafora-body
formation and disease progression.30 of onset and clinical severity.41 Common clinical
manifestations include myopathy, neuropathy, hearing
Myoclonic epilepsy with ragged red fibres loss, dementia, short stature, and optic atrophy. Less
Myoclonic epilepsy with ragged red fibres (MERRF) is one commonly, cardiomyopathy, pigmentary retinopathy,
of the common causes of PME. It may be sporadic or pyramidal signs, ophthalmoparesis, multiple lipomas,
familial. Mitochondrial diseases can be both maternally and diabetes mellitus can occur.42 There is an overlap
and paternally inherited, but typical MERRF, and the with the syndrome of MELAS, but MERRF usually has a
syndrome of mitochondrial encephalomyopathy, lactic longer course and is associated with milder behavioural
acidosis, and stroke-like episodes (known as MELAS), are and cognitive deficits.43
maternally inherited.40 In patients with MERRF, the EEG shows generalised
spike-and-wave discharges at 2–5 Hz, with background
Clinical picture and investigations slowing that progresses as the disease advances. Focal
The syndrome of MERRF is characterised by myoclonus, epileptiform discharges can also be seen.44 Muscle
generalised epilepsy, ataxia, and ragged red fibres in biopsy shows ragged red fibres in over 90% of patients.45
muscle biopsy (figure 2). The clinical spectrum can vary, Biochemical studies of respiratory-chain enzymes in
and the syndrome exhibits intrafamilial variation in age muscle extracts usually show decreased activity.46 Brain

Disease Inheritance Chromosome locus Gene Protein Function Mutation


ULD AR Ch21q22.3 CSTB Cystatin B Cysteine protease inhibitor Unstable expansion of dodecamer repeat or point
mutations
Lafora’s disease AR Ch6q24 EPM2A Laforin Dual specificity protein tyrosine phosphatase More than 20 mutations
Ch6p22 NHLRC1 ··
MERRF Maternal Mitochondrial DNA MTTK tRNALys Mitochondrial function and metabolism Point mutations
NCL
Classical late infantile AR Ch11p15 TPP1 Tripeptidyl peptidase 1 ·· Multiple
Juvenile AR Ch6p CLN3 ·· ·· 1·02 kbp deletion
Adult (Kufs disease) AR/AD ·· ·· ·· ·· ··
Finnish-variant late infantile AR Ch13q21-q32 CLN5 ·· ·· CLN5 Finnish major
Variant late infantile AR Ch15q21-23 CLN6 ·· ·· Multiple
Sialidoses
Type I AR Ch6p21.3 NEU1 Sialidase 1 ·· Multiple
Type II AR Ch20 NEU1 Sialidase 1 ·· Multiple
DRPLA AD Ch12p13.31 DRPLA Atrophin 1 ·· Unstable expansion of CAG repeats

MERRF=myoclonic epilepsy with ragged red fibres; NCL=neuronal ceroid lipofuscinoses; DRPLA=dentatorubral-pallidoluysian atrophy; AR=autosomal recessive; AD=autosomal dominant; ··= unknown.

Table 1: Molecular genetics of PME

http://neurology.thelancet.com Vol 4 April 2005 241


Review

Antigenomic peptide nucleic acids that specifically


inhibit replication of mutant but not wildtype
mitochondrial DNA templates in vitro have been
developed and expressed in cultured human myoblasts.54
Furthermore, mutant mitochondrial DNA predominates
in mature myofibres, but is rare or undetectable in
skeletal muscle satellite cells, a pattern that is thought to
result from positive selection for the mutant
mitochondrial DNA in postmitotic myofibres and loss of
the mutant by genetic drift in satellite cells. This finding
lends itself to new approaches for treatment, for
example, by increasing the incorporation of satellite cells
through regeneration after injury to myofibres by
resistance exercise training,55 or by bupivacaine
hydrochloride injection.56

Neuronal ceroid lipofuscinoses


The neuronal ceroid lipofuscinoses (NCLs) are
characterised by the accumulation of abnormal amounts
Figure 2: Ragged red fibres in the skeletal muscle fibre of a patient with MERRF
of lipopigment in lysosomes (figure 3). There are five
types of NCL that may cause PME: classic late infantile
MRI may show brain atrophy and basal ganglia NCL (type 2) or Jansky-Bielschowsky disease; juvenile
calcifications.43 Grey-matter signal changes on NCL (type 3), Spielmeyer-Vogt-Sjogren disease, or Batten
T2-weighted images are sometimes seen, with deep disease; adult NCL (type 4), Kuf’s disease, or Parry
cerebral nuclei being more involved than the cerebral disease; late infantile Finnish variant NCL (type 5); and
cortex. When signal changes are seen in the white late infantile variant NCL (type 6). Each form of the
matter, the peripheral white matter is the earliest to be disease is genetically distinct, with an autosomal recessive
involved.47 inheritance in all except for the adult form, which may
have autosomal dominant inheritance.57
Genetics and diagnosis
The most common molecular defect is an adenosine to Classic late infantile NCL
guanine substitution at nucleotide pair 8344 (8344A→G) Classic late infantile NCL usually has an onset between
in the tRNALys gene (MTTK) of mitochondrial DNA, and 2·5 and 4 years. Myoclonic, tonic-clonic, atonic, and
is present in 90% of typical MERRF patients.48 Another atypical absence seizures are typically the first
rare identified molecular cause of MERRF is a tyrosine manifestation of the disease. Within a few months from
to cytosine substitution (8356T→C) in the same gene,49 onset, ataxia and psychomotor regression appear,
and another is a guanine to adenosine substitution whereas visual failure develops later. Optic fundi show
(8363G→A).50 However, in some individuals, a mutation attenuated retinal vessels and macular degeneration
has not been identified. Clinical clues to the presence of (table 2). Epilepsy is intractable and dementia and
MERRF include deafness, optic atrophy, myopathy, spasticity are relentlessly progressive, with death
lipomas, intrafamilial variation in age of onset, and occurring about 5 years after onset.25
maternal transmission. Ragged red fibres and mutations EEG shows background slowing and disorganisation
in germline DNA (eg, peripheral blood) can be used to with generalised epileptiform discharges. Specific
confirm the diagnosis.51 electrophysiological features are posterior spikes in
response to low-frequency photic stimulation in EEG
Treatment studies, and the giant visual evoked potentials elicited
There is no specific therapy for MERRF, but, as in many only with flash stimulation.58 The gene for this disease
mitochondrial disorders, many patients are empirically (TPP1) has been mapped to chromosome 11p15.59 This
treated with combinations of antioxidant vitamins and gene encodes the protein tripeptidyl peptidase 1 (TPP1).
cofactors including coenzyme Q10 and L-carnitine.52 Multiple mutations that include missense, nonsense,
Valproic acid causes inhibition of carnitine uptake, so deletion, insertion, and splicing mutations have been
must be used with caution and combined with detected.60 TPP1 removes tripeptides from the
L-carnitine supplementation.53 N-terminal of proteins undergoing degradation in
In most patients, mutant and wildtype mitochondrial lysosomes. The specificity and substrate range of TPP1
DNA molecules coexist (heteroplasmy), and there is a is still not clear. Reduced or undetectable TPP1 enzyme
threshold amount of mutant mitochondrial DNA activity in fibroblasts or leucocytes can be used to
necessary for the disease to be expressed clinically. confirm the diagnosis.58

242 http://neurology.thelancet.com Vol 4 April 2005


Review

Late infantile Finnish variant NCL


A variant of late infantile NCL has been found in Finland
(NCL type 5). This variant differs from classic late
infantile NCL (type 2) in that onset is later, at around age
5 years, and includes symptoms of clumsiness and
hypotonia. This is followed by visual impairment at age
5–7 years, and then ataxia at 7–10 years. Myoclonic and
tonic-clonic seizures usually appear at around 8 years of
age. Progression is slower than in NLC type 2. EEG is
similar to that in NCL type 2, but the substantial
response to photic stimulation develops later, at age
7–8 years.61
The gene associated with the disease, CLN5, is found
almost exclusively in Finland and has been mapped to
chromosome 13q21–q32.62 The most common mutation
(Finnish major mutation or CLN5 Fin major) occurs in
94% of Finnish patients and is a 2 bp deletion in exon 4.
The CLN5 gene encodes for a putative transmembrane
protein of 407 amino acid residues, the function of
which is unknown.58 Figure 3: Section of cortex examined under fluorescent light to show yellow intracytoplasmic autofluorescence
typical of the accumulated lipofuscin in Kuf’s disease
Late infantile variant NCL
A variant of late infantile NCL, sometimes called early reveals optic atrophy, macular degeneration, and
juvenile NCL, Gypsy-Indian late infantile NCL, or NCL attenuated vessels. Death occurs about 8 years after
type 6, features an intermediate onset of symptoms at disease onset.64
age 5–7 years and a course that leads to death in the mid EEG shows a slow background with generalised spike
twenties. The associated gene, CLN6, has been mapped and wave. Epileptiform abnormalities are accentuated
to chromosome 15q21–23. The encoded protein and during sleep but not with photic stimulation.65 The gene
function are unknown.59 There is no major founder associated with this disease, CLN3, is located on the
mutation for this disease; 18 mutations have been short arm of chromosome 16.66,67 The CLN3 gene
reported, including missense, nonsense, small deletions encodes a protein of 438 amino-acid residues, the
or insertions, and splice-site mutations. Cases reported function of which is unknown. The most common
in Costa Rica, Venezuela, Pakistan, and the Czech mutation of more than 25 mutations identified is a
Republic show that families from the same country do 1·02 kbp deletion that removes exons 7 and 8, resulting
not all share the same mutation.63 in production of a truncated protein containing the
original 153 N-terminal residues followed by 28 new
Juvenile NCL amino acid residues.68
Juvenile-onset NCL, also known as Batten disease or
NCL type 3, starts at age 4–10 years with visual failure. Adult NCL
Dementia and extrapyramidal features develop Adult NCL (Kuf’s disease, or NCL type 4), may have its
gradually, and seizures are not a prominent onset in childhood or adolescence, despite its
manifestation of the disease. Most patients are blind by denomination. Myoclonus can first occur as late as age
their second decade. The most common seizure type is 30 years. Dementia, ataxia, and extrapyramidal signs
generalised tonic-clonic; myoclonus is usually subtle. may develop first. There are no ophthalmological
Behavioural and psychiatric problems, including abnormalities or visual failure.69 EEG shows generalised
psychosis and hallucinations are common. Fundoscopy fast spike-and-wave discharges with photosensitivity.70

Disease Age at onset (years) Prominent seizures Cerebellar signs Dementia Fundi Dysmorphism
ULD 6–15 Myoclonus ++++ Mild and late Mild and late or absent Normal No
Lafora’s disease 12–17 Myoclonus and occipital seizures Early Early and relentless Normal No
MERRF Any age Myoclonus ++ Variable Variable With or without optic atrophy or retinopathy With or without
NCL Variable Variable Variable Rapidly progressive Macular degeneration and visual failure, except No
Kuf’s disease
Sialidoses Variable Myoclonus +++ Gradual Absent in type I; learning difficulty in type II Cherry-red spot type II ++

ULD=Unverricht-Lundborg disease; MERRF=myoclonic epilepsy with ragged red fibres; NCL=neuronal ceroid lipofuscinoses; +=prominent feature

Table 2: Some differentiating clinical characteristics of PME

http://neurology.thelancet.com Vol 4 April 2005 243


Review

The disease is sometimes observed as a sporadic case, These strategies are based on the concept that the pro-
or as familial disease with autosomal recessive TPP1 protein is taken up via mannose-6-phosphate
inheritance (most commonly), or with autosomal receptors on both the producer and the neighbouring
dominant inheritance (least commonly), suggesting cells.79,80 Recently, virus-mediated gene transfer of TPP1
genetic heterogeneity of the disease.57 to the CNS of mice has been reported as a potential
therapy for late infantile NCL.81
Diagnosis and treatment of NCL
Diagnosis of NCL is typically suspected clinically Sialidoses
(table 2) and supported by electrophysiological studies. Two sialidoses are rare causes of PME. Sialidoses type I
Visual and ophthalmological abnormalities point to NCL (cherry-red spot myoclonus syndrome) is caused by
forms other than type 4. MRI findings in NCL include deficiency of  neuraminidase. It has a juvenile or adult
cerebral and cerebellar atrophy, T2-hyperintensity of the onset and produces a rather pure intention and action
lobar white matter, and thinning of the cerebral cortex.71 myoclonus. Slow progression and absence of mental
Definitive diagnosis currently relies on finding typical deterioration or dysmorphism are characteristic of the
intracellular inclusions by electron microscopy,72 which syndrome. There is gradual visual failure, tonic-clonic
can be located in eccrine secretory cells and also in seizures, ataxia, and a characteristic cherry-red spot in
conjuctival or muscle biopsy. Suction biopsy of rectal the fundus.82
mucosa is also a reliable method to show the Sialidoses type II is caused by a deficiency of both
inclusions.73 The inclusions have different morphologies N-acetyl neuraminidase and -galactosialidase, and is
that point to individual types of NCL, being curvilinear therefore also called galactosialidoses. The time of onset
in late infantile NCL and fingerprint figure in juvenile varies from the neonatal period to the second decade of
and adult NCL (figure 4).74–76 life. Clinical features include coarse facial features,
There is no specific treatment and like most other corneal clouding, hepatomegaly, skeletal dysplasia, and
causes of PME the mainstay of treatment is supportive learning disability in addition to the myoclonus. EEG
and palliative care. Antioxidant treatment in patients background shows low-voltage fast activity, but slowing
with juvenile NCL, with vitamins E and C and can be seen in patients with dementia. Massive
methionine, has been tried in one study but a definite myoclonus is associated with trains of 10–20 Hz, small,
benefit has not been confirmed.77 Gene therapy is still vertex-positive spikes preceding the electromyographic
experimental. Treatment options for the future include artefact.83 MRI findings in sialidoses range from normal
delivering active TPP1 to CNS and retinal cells in a in the early stages to cerebellar, pontine, and cerebral
sufficient quantity and time to prevent cell loss. atrophy as the disease progresses.84
Theoretically, this can be achieved by enzyme The sialidoses are autosomal recessive disorders. The
augmentation therapy, allogenic stem-cell human sialidase gene (NEU1) is located inside the locus
transplantation, and other forms of gene therapy.78 of the MHC on chromosome 6p21.3.85 Sialidase is
targeted to the endosomal-lysosomal compartment as an
integral membrane protein by vesicular transport, which
involves association of the adapter proteins with a
tyrosine-containing internalisation signal at the
C-terminus of the enzyme.86 There are no major founder
mutations for the sialidoses. Several mutations
including five novel mutations have been described; the
mutated protein may prevent substrate binding or
impair the folding of the sialidase enzyme.87 More than
39 mutations in NEU1 have been described,87,88
including splice-site mutations, insertions and deletions,
nonsense, and missense mutations. The most common
of these are the missense mutations, which cause almost
complete loss of enzyme activity. The analysis of these
mutations reveals substantial molecular heterogeneity,
reflecting the diversity of clinical phenotypes. Patients
with a combination of a mild and a severe mutation have
a clinically milder form of the disease, suggesting that a
small percentage of normal sialidase activity may protect
against severe phenotypes. Hence, enzyme replacement
therapy is a possible approach to treatment in
Figure 4: Fingerprint intraneuronal inclusion typical of Kuf’s disease (electron micrograph) sialidoses.88 The gene responsible for galactosialidoses is
Reproduced with permission from Dustri Verlag.76 located on chromosome 20.89 Protective protein

244 http://neurology.thelancet.com Vol 4 April 2005


Review

cathepsin A (PPCA), a lysosomal carboxypeptidase, is myoclonus, dysarthria, ataxia, and renal failure.101 A
deficient in galactosialidoses.90 Enzyme-replacement juvenile form of Huntington’s disease may also cause
experiments have been done on PPCA-deficient mice, PME.102 Familial encephalopathy with neuroserpin
which display symptoms similar to those of severe inclusion bodies is a rare autosomal-dominant disease
galactosialidoses. These mice received haematopoietic that causes progressive dementia and, in some cases, a
progenitors transduced with a bicistronic retroviral familial form of PME. It is caused by mutation in the
vector based on a murine stem-cell virus, which gene coding for the serine protease inhibitor (serpin) on
overexpresses PPCA and a fluorescent protein marker. chromosome 3q26. Inclusion bodies are distributed
Complete correction of the disease phenotype was seen throughout the cerebral cortex and substantia nigra.103
up to 10 months after transplantation, and PPCA- Non-infantile neuronopathic Gaucher’s disease,
positive cells derived from the bone marrow were atypical inclusion body disease, neuraxonal dystrophy,
detected in all tissue including the brain.91 The cherry- coeliac disease, juvenile GM2 gangliosidosis,
red spot should be sought when sialidoses is suspected Hallervorden-Spatz disease, and Alzheimer’s disease
clinically. Diagnosis is confirmed by the detection of beginning in the third or fourth decade are other very
high urinary sialyloligosaccharides and by confirmation rare causes of a PME phenotype.104
of the lysosomal enzyme deficiency in leucocytes or
cultured fibroblasts.92 Conclusion: diagnosis and treatment of PME
The PMEs are rare disorders, that typically have a
Dentatorubral-pallidoluysian atrophy genetic cause. Most PMEs are autosomal-recessive
Dentatorubral-pallidoluysian atrophy (DRPLA) is a rare disorders, except for adult NCL (Kuf’s disease), DRPLA,
autosomal-dominant neurodegenerative disorder, juvenile Huntington’s disease, familial encephalopathy
characterised by various combinations of cerebellar with neuroserpin inclusion bodies, and MERRF. The
ataxia, choreoathetosis, myoclonus, epilepsy, dementia, mode of inheritance, especially if dominant, can give a
and psychiatric symptoms. First described in Japan, clue to diagnosis.
where it is most prevalent, molecular diagnosis has In the early stages of PME, clinical and EEG features
allowed detection of the disease in other ethnic may mimic idiopathic generalised epilepsy syndromes,
groups.93–95 There is substantial heterogeneity in clinical especially juvenile myoclonic epilepsy, but failure of
presentation, even in the same family. There are three therapy and progressive neurological and EEG
clinical forms: an ataxochoreoathetoid form, a pseudo- deterioration point to a diagnosis of PME. Conversely,
Huntington form, and a PME form. Patients with onset the clinical picture of patients with idiopathic
before age 20 years often present with the phenotype of generalised epilepsy may mimic PME, if they are
PME, characterised by ataxia, seizures, myoclonus, and inappropriately treated and intoxicated with antiepileptic
progressive intellectual deterioration.96,97 drugs, with resultant ataxia, impaired cognitive function,
DRPLA is an autosomal-dominant disorder, caused by and poorly controlled seizures.1 However,
unstable expansion of CAG repeats of a gene at photosensitivity may require photic stimulation at a
12p13.31.98,99 The discovery of the DRPLA gene has made lower frequency than that in idiopathic generalised
it possible to analyse the diverse clinical presentations on epilepsy. A full history of the illness, developmental
the basis of the size of the expanded CAG repeats. There history in children, comprehensive family history when
is also an inverse correlation between the age at onset possible, and thorough clinical examination are
and the size of the expanded CAG repeats. DRPLA is imperative to obtain clues to diagnosis.
characterised by prominent anticipation with a mean Although laboratory and pathological studies are still
acceleration of age at onset of 26·5 years (SD 2·4) in required for some of the PME disorders, the revolution
paternal transmission and 14 years (SD 4) in maternal in molecular genetics has allowed a definitive diagnosis
transmission. MRI findings include atrophy of mid- of some PMEs such as Unverricht-Lundborg disease,
saggital structures of the cerebellum and brain stem, MERRF, DRPLA, and most patients with Lafora’s
particularly the pontine tegmentum. There is strong disease. Reaching a definite diagnosis has significant
inverse correlation between the age at diagnosis by MRI implications for management and genetic counselling.
and the areas of atrophy in patients with large expanded Treatment of PME disorders remains essentially that
CAG repeats. However, cerebral white-matter of managing seizures and myoclonus together with
involvement is associated with the duration of the illness palliative, supportive, and rehabilitative measures.
rather than with the size of the CAG repeats.100 Diagnosis Treatment of myoclonus and seizures in PME can prove
is confirmed by identifying the abnormal CAG repeats. to be difficult as both tend to be refractory and resistant
to conventional antiepileptic medications. Controlled
Other rare causes of PME clinical studies could be used to assess the antiepileptic
The action-myoclonus renal-failure syndrome, an drug efficacy in PME in large groups of patients, but are
autosomal recessive disorder, was described in French not possible because the incidence of these disorders is
Canadians with PME, severe progressive action rare. Hence, available data on the efficacy of newer

http://neurology.thelancet.com Vol 4 April 2005 245


Review

Authors’ contributions
Panel 2: Drugs and PME AS designed and wrote the review and collected the data. MF revised the
review and provided the neuropathological figures. ND developed the
Used for treatment initial idea, contributed to the design of the review, and revised the
paper at its different stages of development for important intellectual
Valproic acid
content.
Benzodiazepines
Phenobarbital Conflicts of interest
ND has received unrestricted educational and research grant support
Piracetam
from the following pharmaceutical companies: UCB Pharma, Elan
Levetiracetam Pharma, GlaxoSmithKline, Jannsen Cilag, Pfizer and Novartis. He is or
Zonisamide has also been a member of Elan Advisory Board (Ireland), UCB Pharma
Advisory Board (UK), Eisai Advisory Board (Europe), Irish advisory
May aggravate myoclonus boards of Janssen Cilag and GlaxoSmithKline, and the European
Phenytoin advisory board of Pfizer. He has also received speaker’s honoraria from
Carbamazepine UCB Pharma and Jannsen Cilag. The decision to write and submit this
review was entirely his, and was not in any way influenced or initiated
Gabapentin by any pharmaceutical company. MF is not employed by any
Vigabatrin pharmaceutical companies nor does he receive any funds from
Tiagabine pharmaceutical or other commercial companies, nor does he have
ownership of stocks in such companies. AS is not employed by any
Use with caution pharmaceutical company nor does he receive any funds from
Lamotrigine in all cases pharmaceutical or commercial companies, nor does he have ownership
of stocks in such companies.
Valproic acid in MERRF
Role of the funding source
There is no funding allocated for this study and it is entirely the sole
antiepileptic medications in PME are primarily efforts of the participating authors.
anecdotal or observational, based on responses of
References
individuals or very small groups of patients.105 1 Berkovic SF. Progressive myoclonic epilepsies. In: Engel J, Pedley
Commonly used antiepileptic drugs for management TA, eds. Epilepsy: a comprehensive textbook. Philadelphia:
of myoclonus (panel 2) include combinations of valproic Lippincott-Raven, 1997: 2455–68.
2 Unverricht H. Die Myoclonie. Leipzig: Franz Deuticke, 1891.
acid, benzodiazepines, phenobarbital, and more 3 Lundborg H. Die progressive Myoclonus-Epilepsie (Unverricht’s
recently, piracetam, zonisamide, and levetiracetam. Myoclonie). Uppsala: Almqvist and Wiksell, 1903.
Vagus-nerve stimulation may also have a role but needs 4 Norio R, Koskiniemi M. Progressive myoclonus epilepsy: genetic
and nosological aspects with special reference to 107 Finish
to be further investigated and can not yet be considered patients. Clin Genet 1979; 15: 382–98.
as a routine treatment. Care must be taken to avoid 5 Genton P, Michelucci R, Tassinari CA, Roger J. The Ramsay Hunt
antiepileptic medications that clearly worsen syndrome revisited: Mediterranean myoclonus versus
mitochondrial encephalomyopathy with ragged-red fibers and
myoclonus. These include vigabatrin, carbamazepine, Baltic myoclonus. Acta Neurol Scand 1990; 81: 8–15.
phenytoin, and gabapentin. Lamotrogine has an 6 Koskiniemi M, Donner M, Majuri H, Haltia M, Norio R.
unpredictable effect on myoclonus and must be used Progressive myoclonus epilepsy: a clinical and histopathologic
study. Acta Neurol Scand 1974; 50: 307–32.
with caution.106–108
7 Lehesjoki AE. Clinical features and genetics of Unverricht-
Through recent advances in molecular genetics, Lundborg disease. Adv Neurol 2002; 89: 193–97.
several gene loci, mutations, and proteins involved in the 8 Kyllerman M, Sommerfelt TK, Hedstrom A, Wennergren G,
pathogenesis of PME disorders have been identified. Holmgren D. Clinical and neurophysiological development of
Unverricht-Lundborg disease in four Swedish siblings. Epilepsia
Future treatments with gene therapy and enzyme 1991; 32: 900–09.
replacement may help to modify and improve the course 9 Kinirons P, Ibrahim N, Murphy K, Lehesjoki AE, Jarvela I, Delanty
of these progressive disorders. N. Efficacy of levetiracetam in a patient with Unverricht-Lundborg
progressive myoclonic epilepsy. Neurology 2003; 60: 1394.
10 Berkovic SF, So NK, Andermann F. Progressive myoclonus
epilepsies: clinical and neurophysiological diagnosis.
Search strategy and selection criteria J Clin Neurophysiol 1991; 8: 261–74.
11 Mascalchi M, Michelucci R, Cosottini M, et al. Brainstem
Data for this review were identified by searches of PubMed with involvement in Unverricht-Lundborg disease (EPM1): an MRI and
the terms “myoclonus”, “progressive myoclonic epilepsy”, 1
H MRS study. Neurology 2002; 58: 1686–89.
“Unverricht-Lundborg disease”, “Lafora disease”, “myoclonic 12 Lehesjoki AE, Koskiniemi M, Sistonen P, et al. Localization of a
gene for progressive myoclonus epilepsy to chromosome 21q22.
epilepsy with ragged red fibres”, “neuronal ceroid Proc Natl Acad Sci U S A 1991; 88: 3696–99.
lipofuscinosis”, “sialidoses”, “dentatorubral-pallidoluysian 13 Virtaneva K, Miao J, Traskelin AL. Progressive myoclonus epilepsy
atrophy”, “genetics”, “mutations”, and “myoclonus treatment”. EPM1 locus maps to a 175 kb interval in distal 21q.
Am J Hum Genet 1996; 58: 1247–53.
References and textbooks were also identified through 14 Pennacchio LA, Lehesjoki AE, Stone NE, et al. Mutations in the
references cited in relevant articles. Only papers published in gene encoding cystatin B in progressive myoclonus epilepsy
English were reviewed. The search covered publications from (EPM1). Science 1996; 271: 1731–34.
15 Lafreniere RG, Rochefort DL, Chretien N, et al. Unstable
January, 1971, to November, 2004, with inclusion of two insertion in the 5´-flanking region of the cystatin-B gene is the
historical references from 1891 and 1903. most common mutation in progressive myoclonus epilepsy type-1,
EPM1. Nat Genet 1997; 15: 298–302.

246 http://neurology.thelancet.com Vol 4 April 2005


Review

16 Lalioti MD, Mirotsou M, Buresi C, et al. Identification of mutations 40 Rosing HS, Hopkins LC, Wallace DC, Epstein CM, Weidenheim K.
in cystatin B, the gene responsible for the Unverricht-Lundborg Maternally inherited mitochondrial myopathy and myoclonic
type of progressive myoclonus epilepsy (EPM1). Am J Hum Genet epilepsy. Ann Neurol 1985; 17: 228–37.
1997; 60: 342–51. 41 Berkovic SF, Carpenter S, Evans A, et al. Myoclonus epilepsy and
17 Delgado-Escueta AV, Ganesh S, Yamakawa K. Advances in the ragged-red fibers (MERRF). 1. A clinical, pathological, biochemical,
genetics of progressive myoclonus epilepsy. Am J Med Genet 2001; magnetic resonance spectroscopic and positron emission
106: 129–38. tomographic study. Brain 1989; 112: 1231–60.
18 Di Giaimo R, Riccio M, Santi S, Galeotti C, Ambrosetti DC, Melli M. 42 Chinnery PF, Howell N, Lightowlers RN, Turnbull DM.
New insights into the molecular basis of progressive myoclonus Molecular pathology of MELAS and MERRF. The relationship
epilepsy: a multi-protein complex with cystatin B. Hum Mol Genet between mutation load and clinical phenotypes. Brain 1997;
2002; 11: 2941–50. 120: 1713–21.
19 Iivanainen M, Himberg JJ. Valproate and clonazepam in the 43 DiMauro S, Hirano M, Kaufmann P, et al. Clinical features and
treatment of severe progressive myoclonus epilepsy. Arch Neurol genetics of myoclonic epilepsy with ragged-red fibers. Adv Neurol
1982; 39: 236–38. 2002; 89: 217–29.
20 Remy C, Genton P. Effect of high dose of oral piracetam on 44 So N, Berkovic SF, Andermann F, Kuzniecky R, Gendron D,
myoclonus in progressive myoclonus epilepsy (Mediterranean Quesney LF. Myoclonus epilepsy and ragged-red fibers (MERRF).
myoclonus). Epilepsia 1991; 32: 6. 2. Electrophysiological studies and comparison with the other
21 Magaudda A, Gelisse P, Genton P. Antimyoclonic effect of progressive myoclonus epilepsies. Brain 1989; 112: 1261–76.
levetiracetam in 13 patients with Unverricht-Lundborg disease: 45 Hirano M, DiMauro S. Clinical features of mitochondrial
clinical observations. Epilepsia 2004; 45: 678–81. myopathies and encephalomyopathies. In: Lane RJM, ed.
22 Henry TR, Leppik IE, Gumnit RJ, Jacobs M. Progressive Handbook of muscle disease. New York: Marcel Dekker Inc., 1996:
myoclonus epilepsy treated with zonisamide. Neurology 1988; 479–504.
38: 928–31. 46 Boulet L, Karpati G, Shoubridge EA. Distribution and threshold
23 Eldridge R, Iivanainen M, Stern R, Koerber T, Wilder BJ. “Baltic” expression of the tRNALys mutation in skeletal muscle of patients
myoclonus epilepsy: hereditary disorder of childhood made worse with myoclonic epilepsy and ragged-red fibers (MERRF).
by phenytoin. Lancet 1983; ii: 838–42. Am J Hum Genet 1992; 51: 1187–200.
24 Smith B, Shatz R, Elisevich K, Bespalova IN, Burmeister M. 47 Barkovich AJ, Good WV, Koch TK, Berg BO. Mitochondrial
Effects of vagus nerve stimulation on progressive myoclonus disorders: analysis of their clinical and imaging characteristics.
epilepsy of Unverricht-Lundborg type. Epilepsia 2000; 41: 1046–48. Am J Neuroradiol 1993; 14: 1119–37.
25 Berkovic SF, Andermann F. The progressive myoclonus epilepsies. 48 Shoffner JM, Lott MT, Lezza A, Seibel P, Ballinger SW, Wallace DC.
In: Pedley TA, Meldrum BS, eds. Recent advances in epilepsy 3. Myoclonic epilepsy and ragged-red fiber disease (MERRF) is
Edinburgh: Churchill Livingstone, 1986: 157–87. associated with a mitochondrial DNA tRNALys mutation. Cell 1990;
26 Busard BLSM, Renier WO, Gabreels FJM, Jaspar HHJ, 61: 931–37.
van Haelst UJG, Stooff JL. Lafora’s disease. Arch Neurol 1986; 49 Silvestri G, Moraes CT, Shanske S, Oh SJ, DiMauro S. A new
43: 296–99. mtDNA mutation in the tRNALys gene associated with myoclonic
27 Acharya JN, Satishchandra P, Shankar SK. Familial progressive epilepsy and ragged-red fibers (MERRF). Am J Hum Genet 1992;
myoclonus epilepsy: clinical and electrophysiologic observations. 51: 1213–17.
Epilepsia 1995; 36: 429–34. 50 Santorelli FM, Mak SC, El-Schahawi M, et al. Maternally inherited
28 Kobayashi K, Iyoda K, Ohtsuka Y, Ohtahara S, Yamada M. cardiomyopathy and hearing loss associated with a novel mutation
Longitudinal clinicoelectrophysiologic study of a case of Lafora in the mitochondrial DNA tRNALys gene (G8363A).
disease proven by skin biopsy. Epilepsia 1990; 31: 194–201. Am J Hum Genet 1996; 58: 933–39.
29 Berkovic SF, Cochius J, Andermann E, Andermann F. Progressive 51 Hammans SR, Sweeney MG, Brockington M, Morgan-Hughes JA,
myoclonus epilepsies: clinical and genetic aspects. Epilepsia 1993; Harding AE. Mitochondrial encephalopathies: molecular genetic
34 (suppl 3): S19–30. diagnosis from blood samples. Lancet 1991; 337: 1311–13.
30 Minassian BA. Progressive myoclonus epilepsy with polyglucosan 52 DiMauro S, Hirano M, Schon EA. Mitochondrial
bodies: Lafora disease. Adv Neurol 2002; 89: 199–210. encephalomyopathies: therapeutic approaches. Neurol Sci 2000;
31 Van Heycop Ten Ham MW. Lafora disease, a form of progressive 21 (suppl 5): S901–08.
myoclonus epilepsy. In: Magnus O, de Haas AML, eds. Handbook 53 Tein I, DiMauro S, Xie Z-W, De Vivo DC. Valproic acid impairs
of clinical neurology: the epilepsies, vol 15. Amsterdam: North carnitine uptake in cultured human skin fibroblasts: an in vitro
Holland Publishing Company, 1974: 382–422. model for pathogenesis of valproic acid-associated carnitine
32 Kobayashi K, Iyoda K, Ohtsuka Y, Ohtahara S, Yamada M. deficiency. Pediatr Res 1993; 34: 281–87.
Longitudinal clinicoelectrophysiologic study of a case of Lafora 54 Taylor RW, Chinnery PF, Turnbull DM, Lightowlers RN. Selective
disease proven by skin biopsy. Epilepsia 1990; 31: 194–201. inhibition of mutant human mitochondrial DNA replication in
33 Roger J, Genton P, Bureau M, Dravet C. Progressive myoclonus vitro by peptide nucleic acids. Nat Genet 1997; 51: 212–15.
epilepsies in childhood and adolescence. In: Roger J, Bureau M, 55 Taivassalo T, Fu K, Johns T, Arnold D, Karpati G, Shoubridge EA.
Dravet C, Dreifus FE, Perret A, Wolf P, eds. Epileptic syndromes in Gene shifting: a novel therapy for mitochondrial myopathy.
infancy, childhood and adolescence, 2nd edn. London: John Libbey, Hum Mol Genet 1999; 8: 1047–52.
1992: 381–400. 56 Andrews RM, Griffiths PG, Chinnery PF, Turnbull DM. Evaluation
34 Yen C, Beydoun A, Drury I. Longitudinal EEG studies in a kindred of bupivacaine-induced muscle regeneration in the treatment of
with Lafora disease. Epilepsia 1991; 32: 895–99. ptosis in patients with chronic progressive external ophthalmoplegia
35 Carpenter S, Karpati G. Sweat gland duct cells in Lafora disease: and Kearns-Sayre syndrome. Eye 1999; 13: 769–72.
diagnosis by skin biopsy. Neurology 1981; 31: 1564–68. 57 Boehme DH, Cottrell JC, Leonberg SC, Zeman W. A dominant
36 Minassian BA, Lee JR, Herbrick JA, et al. Mutations in a gene form of neuronal ceroid-lipofuscinosis. Brain 1971; 94: 745–60.
encoding a novel protein tyrosine phosphatase cause progressive 58 Gardiner RM. Clinical features and molecular genetics basis of the
myoclonus epilepsy. Nat Genet 1998; 20: 171–74. neuronal ceroid lipofuscinoses. Adv Neurol 2002; 89: 211–15.
37 Ganesh S, Agarwala KL, Ueda K, et al. Laforin, defective in 59 Sharp JD, Wheeler RB, Lake BD, et al. Loci for classical and a
progressive myoclonus epilepsy of Lafora type, is a dual specificity variant late infantile neuronal ceroid lipofuscinosis map to
phosphatase associated with polyribosomes. Hum Mol Genet 2000; chromosomes 11p15 and 15q21-23. Hum Mol Genet 1997;
9: 2251–61. 6: 591–95.
38 Chan EM, Bulman DE, Paterson AD, et al. Genetic mapping of a 60 Sleat DE, Gin RM, Sohar I, et al. Mutation analysis of the defective
new Lafora progressive myoclonus epilepsy locus (EPM2B) on protease in classic late-infantile neuronal ceroid lipofuscinosis, a
6p22. J Med Genet 2003; 40: 671–75. neurodegenerative lysosomal storage disorder. Am J Hum Genet
39 Chan EM, Omer S, Ahmed M, et al. Progressive myoclonus 1999; 64: 1511–23.
epilepsy with polyglucosans (Lafora disease). Evidence for a third 61 Santavuori P, Rapola J, Sainio K, Raitta C. A variant of Jansky-
locus. Neurology 2004; 63: 565–67. Bielschowsky disease. Neuropediatrics 1982; 13: 135–41.

http://neurology.thelancet.com Vol 4 April 2005 247


Review

62 Savukoski M, Kestila M, Williams R, et al. Defined chromosomal 86 Lukong KE, Landry K, Elsliger MA, et al. Mutations in sialidoses
assignment of CLN5 demonstrates that at least four genetic loci are impair sialidase binding to the lysosomal multienzyme complex.
involved in the pathogenesis of human ceroid lipofuscinoses. J Biol Chem 2001; 276: 17286–90.
Am J Hum Genet 1994; 55: 695–701. 87 Pattison S, Pankarican M, Rupar CA, Graham FL, Igdoura SA. Five
63 Sharp JD, Wheeler RB, Parker KA, Gardiner RM, Williams RE, novel mutations in the lysosomal sialidase gene (NEU1) in type II
Mol SE. Spectrum of CLN6 mutations in variant late infantile sialidosis patients and assessment of their impact on enzyme
neuronal ceroid lipofuscinosis. Hum Mutat 2003; 22: 35–42. activity and intracellular target using adenovirus-mediated
64 Rapin I. Myoclonus in neuronal storage and Lafora diseases. expression. Hum Mutat 2004; 23: 32–39.
Adv Neurol 1986; 43: 65–85. 88 Seyrantepe V, Poupetova H, Froissart R, Zabot MT, Maire I,
65 Westmoreland BF, Groover RV, Sharbrough FW. Electrographic Pshezhetsky AV. Molecular pathology of NEU1 gene in sialidosis.
findings in three types of cerebromacular degeneration. Hum Mutat 2003; 22: 343–52.
Mayo Clin Proc 1979; 54: 12–21. 89 Rothschild CB, Akots G, Hayworth R, et al. A genetic map of
66 Eiberg H, Gardiner RM, Mohr J. Batten disease (Spielmeyer- chromosome 20q12-q13.2. Am J Hum Genet 1993; 52: 110–23.
Sjogren disease) and haptoglobins (HP): indication of linkage and 90 Galjart NJ, Gillemans N, Harris A, et al. Expression of cDNA
assignment to chromosome 16. Clin Genet 1989; 36: 217–18. encoding the human “protective protein” associated with lysosomal
67 Jarvela IE, Mitchison HM, Callen DF. Physical map of the region beta-galactosidase and neuraminidase: homology to yeast
containing the gene for Batten disease (CLN3). Am J Med Genet proteases. Cell 1988; 54: 755–64.
1995; 57: 316–19. 91 Leimig T, Mann L, Martin MdP, et al. Functional amelioration of
68 Haskell RE, Carr CJ, Pearce DA, Bennett MJ, Davidson BL. Batten murine galactosialidosis by genetically modified bone marrow
disease: evaluation of CLN3 mutations on protein localization and hematopoietic progenitor cells. Blood 2002; 99: 3169–78.
function. Hum Mol Genet 2000; 9: 735–44. 92 Lowden JA, O’Brien JS. Sialidosis: a review of human
69 Berkovic SF, Carpenter S, Andermann F, et al. Kuf’s disease: a neuraminidase deficiency. Am J Hum Genet 1979; 31: 1–18.
critical reappraisal. Brain 1988; 111: 27–62. 93 Warner TT, Williams L, Harding A. DRPLA in Europe. Nat Genet
70 Barthez-Carpentier MA, Billard C, Maheut J, Santini JJ, 1994; 6: 225.
Ruchoux MM. A case of childhood Kuf’s disease. 94 Potter NT, Meyer MA, Zimmerman AW, Eisentadt ML, Anderson IJ.
J Neurol Neurosurg Psychiatry 1991; 54: 655–57. Molecular and clinical findings in a family with dentatorubral-
71 D’Incerti L. MRI in neuronal ceroid lipofuscinosis. Neurol Sci 2000; pallidoluysian atrophy. Ann Neurol 1995; 37: 273–77.
21 (suppl 3): 571–73. 95 Connatry M, Dennis NR, Patch C, Macpherson JN, Harvey JF.
72 Wisniewski KE, Zhong N, Philippart M. Pheno/genotypic Molecular reinvestigation of patients with Huntington’s disease in
correlations of neuronal ceroid lipofuscinoses. Neurology 2001; Wessex reveals a family with dentatorubral and pallidoluysian
57: 576–81. atrophy. Hum Genet 1996; 97: 76–78.
73 Rapola J, Santavuori P, Savilahti E. Suction biopsy of rectal mucosa 96 Natio H, Oyanagi S. Familial myoclonus epilepsy and
in the diagnosis of infantile and juvenile types of neuronal ceroid choreoathetosis: hereditary dentatorubral-pallidoluysian atrophy.
lipofuscinoses. Hum Pathol 1984; 15: 352–60. Neurology 1982; 32: 798–807.
74 Carpenter S. Morphological diagnosis and misdiagnosis in Batten- 97 Iizuka R, Hirayama K, Maehara K. Dentato-rubro-pallido-luysian
Kufs disease. Am J Med Genet Suppl 1988; 5: 85–91. atrophy: a clinico-pathological study. J Neurol Neurosurg Psychiatry
75 Carpenter S, Karpati G, Andermann F, Jacob JC, Andermann E. 1984; 47: 1288–98.
The ultrastructural characteristics of the abnormal cytosomes in 98 Koide R, Ikeuchi T, Onodera O, et al. Unstable expansion of CAG
Batten-Kufs disease. Brain 1977; 100: 137–56. repeat in hereditary dentatorubral-pallidoluysian atrophy (DRPLA).
76 Callagy C, O’Neill G, Murphy SF, Farrell MA. Adult neuronal Nat Genet 1994; 6: 9–13.
ceroid lipofuscinosis (Kufs’ disease) in two siblings of an Irish 99 Nagafuchi S, Yanagisawa H, Sato K, et al. Expansion of an unstable
family. Clin Neuropathol 2000; 19: 109–18. CAG trinucleotide on chromosome 12p in dentatorubral and
77 Santavuori P, Westermark T, Rapola J, et al. Antioxidant treatment pallidoluysian atrophy. Nat Genet 1994; 6: 14–18.
in Spielmeyer-Sjogren’s disease. Acta Neurol Scand 1985; 100 Tsuji S. Dentatorubral-pallidoluysian atrophy: clinical aspects and
71: 136–45. molecular genetics. Adv Neurol 2002; 89: 231–39.
78 Sondhi D, Hackett NR, Apblett RL, Kaminsky SM, Pergolizzi RG, 101 Andermann F, Andermann E, Carpenter S, et al. Action
Crystal RG. Feasibility of gene therapy for late neuronal ceroid myoclonus-renal failure syndrome. Adv Neurol 1986; 43: 87–103.
lipofuscinosis. Arch Neurol 2001; 58: 1793–98. 102 Gambardella A, Muglia M, Labate A, et al. Juvenile Huntington’s
79 Neufeld EF, Fratantoni JC. Inborn errors of mucopolysaccharide disease presenting as progressive myoclonic epilepsy. Neurology
metabolism. Science 1970; 169: 141–46. 2001; 57: 708–11.
80 Taylor RM, Wolfe JH. Cross correction of -glucoronidase 103 Daucher RL, Holohan PD, Shrimpton AE, et al. Familial
deficiency by retroviral vector-mediated gene transfer. Exp Cell Res encephalopathy with neuroserpin inclusion bodies. Am J Pathol
1995; 214: 606–13. 1999; 155: 1901–13.
81 Haskell RE, Hughes SM, Chiorini JA, Alisky JM, Davidson BL. 104 Berkovic SF, Cochius J, Andermann E, Andermann F. Progressive
Viral-mediated delivery of the late-infantile neuronal ceroid myoclonus epilepsies: clinical and genetic aspects. Epilepsia 1993;
lipofuscinosis gene, TPP-I to the mouse central nervous system. 34 (suppl 3): S19–30.
Gene Ther 2003; 10: 34–42. 105 Conry JA. Progressive myoclonic epilepsies. J Child Neurol 2002;
82 Rapin I, Goldfisher S, Katzman R, Engel J Jr, O’Brien JS. The 17 (suppl 1): S80–84.
cherry-red spot myoclonus syndrome. Ann Neurol 1978; 3: 234–42. 106 Shorvon SD. General principles of treatment in epilepsy. In:
83 Engel J Jr, Rapin I, Giblin D. Electrophysiological studies in two Handbook of epilepsy treatment. Oxford: Blackwell Science, 2000:
patients with cherry-red spot myoclonus syndrome. Epilepsia 1977; 71.
18: 73–87. 107 Janszky J, Rasonyi G, Halasz P, et al. Disabling erratic myoclonus
84 Palmeri S, Villanova M, Malandrini A, et al. Type I sialidoses: a during lamotrigine therapy with high serum level, report of two
clinical, biochemical and neuroradiological study. Eur Neurol 2000; cases. Clin Neuropharmacol 2000; 23: 86–89.
43: 88–94. 108 Das KB, Harris C, Smyth DP, Cross JH. Unusual side effects of
85 Pshezhetsky AV, Richard C, Michaud L, et al. Cloning, expression lamotrigine therapy. J Child Neurol 2003; 18: 479–80.
and chromosomal mapping of lysosomal sialidase and
characterization of mutations in sialidosis. Nat Genet 1997;
15: 316–20.

248 http://neurology.thelancet.com Vol 4 April 2005

You might also like