Skip to main content
Erschienen in: Journal of Neurology 7/2015

Open Access 01.07.2015 | Review

Diagnosis, investigation and management of hereditary spastic paraplegias in the era of next-generation sequencing

verfasst von: Anke Hensiek, Stephen Kirker, Evan Reid

Erschienen in: Journal of Neurology | Ausgabe 7/2015

Abstract

The hereditary spastic paraplegias (HSPs) are a group of genetic conditions in which spastic paralysis of the legs is the principal clinical feature. This is caused by a relatively selective distal axonal degeneration involving the longest axons of the corticospinal tracts. Consequently, these conditions provide an opportunity to identify genes, proteins and cellular pathways that are critical for axonal health. In this review, we will provide a brief overview of the classification, clinical features and genetics of HSP, highlighting selected HSP subtypes (i.e. those associated with thin corpus callosum or cerebellar ataxia) that are of particular clinical interest. We will then discuss appropriate investigation strategies for HSPs, suggesting how these might evolve with the introduction of next-generation sequencing technology. Finally, we will discuss the management of HSP, an area somewhat neglected by HSP research.

Introduction

The hereditary spastic paraplegias (HSPs) are a group of genetic conditions in which spastic paralysis of the legs is the principal clinical feature [1, 2]. In most subtypes of HSP, this is caused by a relatively selective distal axonal degeneration involving the longest axons of the corticospinal tracts [3]. As a group, these conditions are moderately rare, with prevalence estimates in different geographical regions ranging from 0.1 to 9.6 per 105 [4]. However, as they are chronic and sometimes severe conditions that frequently present in childhood and young adult life, they represent a significant disease burden. In addition, they are scientifically important, since they provide an opportunity to identify genes, proteins and cellular pathways that are critical for axonal health.
In this review, we will provide a brief overview of the classification, clinical features and genetics of HSP, highlighting selected HSP subtypes that are of particular clinical interest. We will then discuss appropriate investigation strategies for HSPs, suggesting how these might evolve with the introduction of next-generation sequencing technology. Finally, we will discuss the management of HSP, an area somewhat neglected by HSP research.

Overview of main HSP clinical categories

Traditionally, the hereditary spastic paraplegias are subcategorised into “pure” and “complex” (or “complicated”) subtypes [3]. In northern European populations, pure forms are the most frequent. The clinical features of pure HSP have been thoroughly described in previous research articles and reviews, and diagnostic criteria have been proposed [1, 59]. Briefly, the typical clinical picture is of a slowly progressive, predominantly symmetrical, spastic paraplegia. This is frequently accompanied by minor sensory abnormalities (such as absent vibration sensation) and neurological bladder involvement, but bowel involvement is rare. If seen at all, arm involvement is minimal and does not extend beyond hyper-reflexia and minor weakness (e.g. difficulty unscrewing a tight bottle top); more significant arm involvement should raise the possibility of alternative diagnoses such as primary lateral sclerosis or amyotrophic lateral sclerosis, while bulbar involvement is incompatible with a diagnosis of pure HSP. Age at onset varies from childhood to late adult life. Within many multiplex families it is highly variable, although mutations in certain genes, notably spastic paraplegia gene (SPG) 3A/atlastin1, are predominantly associated with a childhood age at onset.
While pure HSPs present a relatively homogenous clinical picture, complex HSPs are a disparate group of distinct disorders (Table 1) [13]. As most complex HSPs are inherited in an autosomal recessive pattern, they are the commonest forms of HSP in populations where consanguinity is frequent; for example, in a Tunisian series of 38 families, approximately 70 % of families had autosomal recessive complex HSP [10]. However, it is important to recognise that these complex subtypes of HSP are found worldwide, e.g. HSP with thin corpus callosum caused by SPG11 or SPG15 mutations has been described in many populations and is the most common type of complex HSP that we see in our own clinical practice in Cambridge, UK.
Table 1
List of known or probable HSP genes
Gene
Protein
Inherited
OMIM
Comment
P/C*
Clinical features of complex forms
SPG1/L1CAM
L1-cell adhesion molecule
XL
#303350
 
C
1. MASA syndrome, 2. X-linked hydrocephalus, 3. X-linked complicated HSP
SPG2/PLP1
Proteolipid protein 1
XL
#312920
 
P/C
1. Pure HSP, 2. complex HSP, 3. Pelizaeus–Merzbacher disease
SPG3A/ATL1
Atlastin1
AD
#182600
 
P
 
SPG4/SPAST
Spastin
AD
#182601
 
P
 
SPG5A/CYP7B1
25-hydroxycholesterol 7-alpha-hydroxylase
AR
#270800
 
P
 
SPG6/NIPA1
NIPA1
AD
#600363
 
P
 
SPG7
Paraplegin
AR
#607259
 
P/C
Cerebellar ataxia, optic atrophy, deafness, amyotrophy
SPG8/KIAA0196
Strumpellin
AD
#603563
 
P
 
SPG9
AD
%601162
 
C
Cataracts, motor neuropathy, skeletal abnormalities and gastro-oesophageal reflux.
SPG10/KIF5A
Kinesin heavy chain 5A
AD
#604187
 
P/C
Peripheral neuropathy, amyotrophy, mental retardation, parkinsonism
SPG11
Spatacsin
AR
#604360
 
C
HSP with thin corpus callosum. Peripheral neuropathy, intellectual disability, cognitive decline, amyotrophy, pseudobulbar involvement, cerebellar involvement, parkinsonism, dystonia
SPG12/RTN2
Reticulon2
AD
#604805
 
P
 
SPG13/HSPD1
Heat shock 60 kDa protein 1
AD
#605280
 
P
 
SPG14
AR
%605229
 
C
Intellectual disability, motor neuropathy
SPG15
Spastizin
AR
#270700
 
C
HSP with thin corpus callosum. Peripheral neuropathy, intellectual disability, cognitive decline, amyotrophy, pseudobulbar involvement, cerebellar involvement, parkinsonism, dystonia, pigmentary maculopathy (Kjellin syndrome).
SPG16
XLR
%300266
 
C
Spastic quadraplegia, intellectual disability, cerebellar ataxia, optic atrophy, nystagmus, bowel and bladder dysfunction.
SPG17/BSCL2
Seipin
AD
#270685
 
C
Silver syndrome; HSP with distal amyotrophy. Overlaps with HMSN V.
SPG18/ERLIN2
ER lipid raft associated 2
AR
#611225
 
C
Epilepsy, intellectual disability, pseudobulbar palsy, joint contractures
SPG19
 
AD
%607152
 
P
 
SPG20/SPARTIN
Spartin
AR
#275900
 
C
Troyer syndrome: dysarthria, pseudobulbar palsy, intellectual disability, amyotrophy, short stature
SPG21/MAST
Maspradin
AR
#248900
 
C
Mast syndrome: progressive dementia, cerebellar signs, extra-pyramidal involvement, thin corpus callosum.
SPG22
SLC16A2
XLR
#300523
 
C
Allan-Herndon-Dudley syndrome
SPG23
AR
%270750
 
C
Disordered skin pigmentation, peripheral neuropathy
SPG24
AR
%607584
 
P
 
SPG25
AR
%608220
 
C
Disc herniation
SPG26/B4GALNT1
Beta-1,4 N-acetylgalactosaminyltransferase 1
AR
#609195
 
C
Intellectual disability, peripheral neuropathy, dysarthria, cerebellar signs, extrapyramidal involvement, cortical atrophy
SPG27
AR
%609041
 
P
 
SPG28/DDHD1
Phospholipase DDHD1
AR
#609340
 
P/C
Cerebellar eye signs, peripheral neuropathy
SPG29/KIF1A
Kinesin family member 1A
AD
%609727
  
Sensorineural hearing impairment, neonatal hyperbilirubinemia, hiatus hernia
SPG30
AR
#610357
 
C
Cerebellar signs, peripheral neuropathy
SPG31/REEP1
Receptor expression-enhancing protein 1
AD
#610250
 
P
 
SPG32
AR
%611252
 
C
Cognitive impairment, thin corpus callosum, cortical atrophy, cerebellar atrophy, pontine dysraphia.
[SPG33/ZFYVE27]
[Protrudin]
[AD]
#610244
Mutation may not be pathogenic-present in control populations
[P]
 
SPG34
XLR
%300750
 
P
 
SPG35/FA2H
Fatty acid 2-hydroxylase
AR
#612319
 
C
Dysarthria, intellectual decline, leukodystrophy, dystonia, optic atrophy, seizures, cerebellar signs, thin corpus callosum. May cause brain iron accumulation.
SPG36
AD
%613096
 
C
Lower limb sensory changes
SPG37
AD
%611945
 
P
 
SPG38
AD
%612335
 
C
Similar to Silver syndrome (see SPG17)
SPG39/PNPLA6
Neuropathy target esterase
AR
#612020
 
C
Distal amyotrophy, cerebellar atrophy. Allelic with Boucher–Neuhauser syndrome (spinocerebellar ataxia, hypogonadotropic hypogonadism, chorioretinal dystrophy)
SPG40
     
SPG41
AD
%613364
Single family with lod score <3
  
SPG42/SLC33A1
Acetyl-coenzyme A transporter 1
AD
#612539
 
P
 
SPG43/C19orf12
C19orf12
AR
#615043
 
P/C
Upper limb involvement, distal amyotrophy. May also be associated with brain iron accumulation
SPG44/GJC2
Gap junction gamma-2 protein
AR
#613206
 
C
Cerebellar signs, seizures, cognitive impairment, scoliosis, leukodystrophy, thin corpus callosum.
SPG45/NT5C2
Cytosolic purine 5’-nucleotidase
AR
#613162
 
C
Optic atrophy, thin corpus callosum, intellectual disability
SPG46/GBA2
Non-lysosomal glucosylceramidase
AR
#614409
 
C
Cerebellar signs, intellectual impairment, cerebral atrophy, cerebellar atrophy, thin corpus callosum, pseudobulbar involvement, cataracts.
SPG47/AP4B1
AP-4 complex subunit beta-1
AR
#614066
 
C
Neonatal hypotonia, severe intellectual impairment, dysmorphic features, thin corpus callosum.
SPG48/AP5Z1
AP-5 complex subunit zeta-1
AR
#613647
 
C
Urinary incontinence
SPG49/TECPR2
Tectonin beta-propeller repeat-containing protein 2
AR
#615031
 
C
Intellectual impairment, dysmorphic features, cerebral atrophy, cerebellar atrophy, thin corpus callosum
SPG50/AP4M1
AP-4 complex subunit mu-1
AR
#612936
 
C
Neonatal hypotonia, severe intellectual impairment, pseudobulbar signs, microcephaly, cerebellar atrophy
SPG51/AP4E1
AP-4 complex subunit epsilon-1
AR
#613744
 
C
Neonatal hypotonia, severe intellectual impairment,dysmorphic features, seizures, cortical atrophy, cerebellar atrophy, microcephaly
SPG52/AP4S1
AP-4 complex subunit sigma-1
AR
#614067
 
C
Neonatal hypotonia, severe intellectual impairment, microcephaly, dysmorphic features, short stature
SPG53/VPS37A
Vacuolar protein sorting-associated protein 37A
AR
#614898
 
C
Intellectual disability
SPG54/DDHD2
Phospholipase DDHD2
AR
#615033
 
C
Intellectual disability, dysarthria, dysphagia, optic hypoplasia, thin corpus callosum and white matter changes, short stature
SPG55/C12ORF65
C12orf65
AR
#615035
 
C
Peripheral neuropathy, optic atrophy, intellectual disability
SPG56/CYP2U1
Cytochrome P450 2U1
AR
#615030
 
P/C
Upper limb involvement, peripheral neuropathy, intellectual impairment, thin corpus callosum
SPG57/TFG
Protein TFG
AR
#615658
 
C
Optic atrophy, peripheral neuropathy
SPG58/SPAX2/KIF1C
Kinesin-like protein KIF1C
AR
#611302
 
C
Cerebellar ataxia
SPG59/USP8
Ubiquitin carboxyl-terminal hydrolase 8
AR
*603158
VUS
P
 
SPG60/WDR48
WD repeat-containing protein 48
AR
*612167
VUS
C
Nystagmus, peripheral neuropathy, intellectual disability
SPG61/ARL6IP1
ADP-ribosylation factor-like protein 6-interacting protein 1
AR
#615685
 
C
Peripheral neuropathy
SPG62
     
SPG63/AMPD2
AMP deaminase 2
AR
#615686
 
P/C
White matter changes in corpus callosum
SPG64/ENTPD1
Ectonucleoside triphosphate diphosphohydrolase 1
AR
#615683
 
C
Dysarthria, intellectual disability, microcephaly, delayed puberty
SPG65
Duplicate of SPG45
     
SPG66/ARSI
Arylsulfatase I
AR
*610009
VUS
C
Thin corpus callosum, cerebellar hypoplasia, colpocephaly, peripheral neuropathy
SPG67/PGAP1
GPI inositol-deacylase
AR
*611655
VUS
C
Intellectual disability, tremor, absent corpus callosum, defective myelination
SPG68/FLRT1
leucine-rich repeat transmembrane protein FLRT1
AR
*604806
VUS
C
Optic atrophy, peripheral neuropathy
SPG69/WARBM2/RAB3GAP2
Rab3 GTPase-activating protein non-catalytic subunit
AR
*609275
 
C
Warburg micro syndrome, Martsolf syndrome
SPG70/MARS
Methionine-tRNA ligase, cytoplasmic
AR
*156560
VUS
P/C
Mild intellectual disability
SPG71/ZFR
Zinc finger RNA-binding protein
AR
*615635
VUS
C
Thin corpus callosum
SPG72/REEP2
Receptor expression-enhancing protein 2
AR/AD
#615625
 
P
 
Spastic ataxias
      
 SPAX1/VAMP1
Vesicle-associated membrane protein 1
AD
#108600
 
SPAX
 
 SPAX2/SPG58/KIF1C
Kinesin-like protein KIF1C
AR
#611302
 
SPAX
 
 SPAX3/MARS2
Methionine-tRNA ligase, mitochondrial
AR
#611390
 
SPAX
 
 SPAX4/MTPAP
Poly(A) RNA polymerase, mitochondrial
AR
#613672
 
SPAX
 
 SPAX5/AFG3L2
AFG3-like protein 2
AR
#614487
 
SPAX
 
 SPAX6/SACS
Sacsin
AR
#270550
 
SPAX
Spastic ataxia of Charlevoix-Saguenay
 EXOSC3/PCH1B
Exosome complex component RRP40
AR
*606489
Allelic with pontocerebellar hypoplasia
  
Note that sometimes the distinction between pure and complex HSP may be somewhat arbitrary. See OMIM entries (http://​www.​ncbi.​nlm.​nih.​gov/​omim/​) for the appropriate primary literature
AD autosomal dominant, AR autosomal recessive, C complex, HMSN hereditary motor and sensory neuropathy, P pure, SPAX spastic ataxia, VUS variant of uncertain significance

Genetics of HSPs

The HSPs show a remarkable degree of genetic heterogeneity. The chromosomal location of more than 70 SPGs is known, and presently more than 50 of the genes have been fully identified (Table 1). Moreover, additional syndromes such as Warburg Micro syndrome, previously considered distinct, have recently been incorporated under the rubric of HSP by reason of overlapping cellular pathology [11]. This new knowledge has led to an evolution of the classification of HSP, which, while still referring to phenotypic information, is increasingly focussed on the gene involved. In addition, gene identification has resulted in the distinction between pure and complex HSP becoming somewhat blurred as it has become apparent that mutations in the same gene may cause either a pure or complex phenotype. An example of this is the rare identification of mutations in SPG4/SPAST, SPG31/REEP1 or SPG3A/atlastin1, three genes typically associated with pure HSP, in patients who have spastic paraplegia accompanied by cerebellar ataxia or peripheral neuropathy [1214].

Pure HSP phenotype

In northern European populations, autosomal dominant inheritance accounts for the majority of patients from families with pure HSP [15]. Autosomal recessive inheritance accounts for most of the remainder, while clear-cut X-linked inheritance is infrequent. Rarely, gonadal mosaicism for a dominant gene has been described and this can be mistaken for autosomal recessive inheritance [16]. In addition, some patients present without a family history. Potential genetic explanations for this are varied and include singleton autosomal recessive cases, new autosomal dominant mutations, or inherited autosomal dominant mutations with non-penetrance in a parent. The estimated relative frequency of mutations in selected genes associated with pure HSP is given in Table 2. From this table, it can be seen that testing for three genes, SPG4/SPAST, SPG3A/Atlastin1 and SPG31/REEP1, will identify the responsible mutation in approximately 50 % of families with autosomal dominant pure HSP. For SPG4/SPAST, this testing should include methodologies to pick up whole exon deletions, as these are a common mutational class in this subtype of HSP (they have also been described in other forms including SPG3A/Atlastin1 and SPG31/REEP1-HSP) [14, 1719]. For families with an autosomal recessive inheritance pattern, screening SPG5 (CYP7B1) will pick up the causative mutations in approximately 5–10 % of cases. However, the yield from testing each individual additional gene beyond these commoner genes is low. Using traditional “one at a time” sequential gene testing to detect these rare mutations can be very expensive and may not be justifiable for publically funded health services. Next-generation sequencing approaches are currently revolutionising this situation (see below).
Table 2
Reported frequencies of mutations in selected pure HSP genes
Gene
Reported frequency in familial cases
Frequency in sporadic cases
Frequency in unselected cases
SPG4/SPAST
31–47 % of AD-pure HSP [4954]
7–18 % [51, 52, 5557]
17–26 % [51, 56, 58]
SPG3A/ATLASTIN1
8–39 % of AD-pure HSP (studies often comprised families with early onset and screened SPG4 negative) [51, 5961]
Unknown
7 % (after exclusion of SPG4 mutations) [13]
SPG31/REEP1
2–8 % of AD-pure HSP (screened families typically SPG4 and/or SPG3 negative) [6264]
2 % [65]
3–7 % [63, 65]
SPG10/KIF5A
3–5 % (series included families with complicated HSP, typically SPG4 and SPG3 negative). A rare cause of pure HSP [6668]
None detected
Unknown
SPG8/KIAA0196
5–8 % of SPG3A and SPG4-negative AD HSP families [69, 70]
Unknown
Unknown
SPG12/RTN2
Rare [71]
Rare
Rare
SPG6/NIPA1
<1 % of AD-pure HSP [72, 73]
Unknown
Rare [72]
SPG13/HSPD1
Rare
Unknown
Unknown
SPG42/SLC33A1
Unknown (single family only)
Unknown
Unknown
SPG5A/CYP7B1
7 % of AR-pure HSP 1 reported AD family [30]
3 % [30]
Unknown
SPG7
Rare cause of AR-pure HSP
7–12 % [34, 74]
<5 % of unselected AR families [75, 76]

Complex HSP phenotypes

As mentioned above, the complex HSPs are a diverse set of conditions, each with distinctive clinical features and each caused by one or a small set of genes. Here, we will focus on two of the more common presentations, HSP with thin corpus callosum and HSP accompanied by cerebellar ataxia.

HSP with thin corpus callosum

A thin corpus callosum on MRI scanning is a characteristic feature of patients who have autosomal recessive SPG11 and SPG15 mutations. These patients have a variable combination of clinical features which, as well as thin corpus callosum, includes typically childhood or teenage onset spastic paraplegia, learning difficulties with progressive cognitive decline, cerebellar signs, ocular involvement, peripheral motor axonopathy and extrapyramidal features including parkinsonism. SPG11/15 mutations are associated with relatively severe gait involvement and patients often eventually require a wheelchair. It is important to note that a thin corpus callosum is not a universal finding in SPG11/15 HSP (e.g. in large series, it was found in 90 % of SPG11 cases and 60 % of SPG15 patients), and that learning difficulties may be present before the spastic gait develops [2022].
Cell biological studies on the SPG11 and SPG15 proteins have revealed why mutations in these genes lead to a similar clinical phenotype. Both SPG11 and SPG15 proteins interact with a 4-protein complex termed AP5 (adapter protein 5), which localises to small intracellular membrane-bound vesicles called endosomes [23, 24]. Although AP5’s function is not fully understood, it probably acts to sort membrane cargoes away from the endosomal compartment [23, 24]. Intriguingly, recessive mutations in a component of the core AP5 complex (SPG48/AP5Z1) also cause a very similar clinical phenotype to SPG11/15 mutations, with cognitive defects, thin corpus callosum and spastic paraplegia [25]. Thus, HSP with thin corpus callosum is an excellent example of the principle that diseases with similar phenotypes are often caused by mutations in functionally related genes. At least four other subtypes of recessive HSP may also present with thin corpus callosum, SPG21/Maspardin, SPG35/FA2H and SPG46/GBA2 and SPG54/DDHD2 ([25] and see Table 1); it remains to be seen whether any of the encoded proteins are functionally related to the AP5 complex.

HSP with cerebellar ataxia

Patients who present with a mixed cerebellar ataxia and spastic paraplegia phenotype represent a particular diagnostic challenge, as the differential diagnosis is very broad. It can be helpful to first make a decision as to whether spastic paraplegia or cerebellar ataxia is the dominant feature. There are consensus diagnostic pathways for patients with predominant cerebellar ataxia and we will not review these further here [26]. In patients where spastic paraplegia is the predominant feature, it is useful to subdivide into pure and complex subtypes, analogous to the subdivision of HSP.
“Complex” spastic ataxia in which the cerebellar ataxia is a (sometimes variable) feature of a more complex syndromal picture. Such conditions include HSP with thin corpus callosum (SPG11/15 mutations), SPG35/FA2H-associated HSP, Troyer syndrome (SPG20) and Mast syndrome (SPG21), SPG26/B4GALNT1, SPG30/KIF1A, SPG39/PNPLA6 and SPG46/GBA2. This group also contains a variety of rare metabolic conditions (reviewed in [27, 28]), including cerebrotendinous xanthomatosis, triple H syndrome, cerebral folate deficiency, metachromatic leukodystrophy, Type III 3-methylglutaconic aciduria, as well as other conditions such as Alexander disease and vanishing white matter disease. Rare patients with Chediak–Higashi syndrome (caused by mutations in the lysosomal trafficking regulator LYST gene) may also present with spastic paraplegia, cerebellar ataxia and peripheral neuropathy, without the hypopigmentary or immune deficiency typically associated with this condition [29].
“Pure” spastic ataxia There are many patients in whom spastic paraplegia with cerebellar involvement is the exclusive clinical presentation. This can occur in three main scenarios:
(a)
As an unusual presentation of mutations in genes that are classically associated with “pure” hereditary spastic paraplegia, including autosomal dominant SPG4/spastin and SPG31/REEP1 and autosomal recessive SPG5/CYP7B1 [12, 14, 30].
 
(b)
As a presentation of genes that are classically associated with cerebellar ataxia. This would especially include Friedreich ataxia and SCA3, in which pyramidal signs may be the presenting feature [31, 32].
 
(c)
As a presentation of mutations in genes that typically cause a spastic paraplegia/cerebellar ataxia overlap syndrome. These genes are often classified under the spastic ataxia (SPAX) nomenclature. Well-recognised examples include the autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS; SPAX6) and SPG7/paraplegin [33, 34]. They also include VAMP1 (SPAX1), KIF1C gene (SPAX2/SPG58), MARS2 (SPAX3), MTPAP (SPAX4) and AFG3L2 (SPAX5/SCA28) (Table 1) [32, 35].
 

Investigation of a patient with suspected HSP

The traditional approach to investigating a patient with possible HSP is to define the phenotype and inheritance pattern, exclude alternative causes, then attempt to make a definitive molecular genetic diagnosis, where possible using clinical and family history information to focus genetic investigations appropriately.

Definition of phenotype, inheritance and exclusion of other causes

As specific investigations of a patient with suspected HSP depend on their clinical features and inheritance pattern, careful clinical phenotyping of index cases and potentially affected family members is crucial. A detailed medical history should include developmental milestones and a three generational family tree. Affected individuals and ideally, apparently unaffected family members should undergo a clinical neurological examination, to document specific neurological features and identify asymptomatic family members who may have subtle abnormal signs on examination. This is particularly important for apparently sporadic cases or isolated sibships, where positive examination findings in an asymptomatic parent will point towards the probability of autosomal dominant inheritance.
Even in individuals who have typical clinical features of HSP and a positive family history of spastic paraparesis, it is important to consider alternative causes and importantly exclude other treatable familial conditions. In his regard, MR imaging of the brain and spinal cord is crucial and should be undertaken in all sporadic cases and at least one family member of familial presentations. Imaging will not only identify many structural, inflammatory or metabolic abnormalities but is also useful to guide investigations into specific genetic causes (for example, by highlighting atrophy patterns). Note that imaging does have an appreciable false negative rate, for example, it can be normal in certain metabolic or inflammatory conditions, including adrenoleukodystrophy or primary progressive multiple sclerosis.
A baseline metabolic blood screen in the index case could include Vitamins B12 and E, creatine kinase, very long chain fatty acids, white cell enzymes, anti-nuclear antibody and copper levels. However, individuals with more complex clinical features or an unclear family history may require further baseline tests. For example, in presumed sporadic pure HSP, a set of more extensive investigations is appropriate, as an inherited basis for the disease has not been proven. This includes CSF examination (including oligoclonal bands, HTLV serology), Syphilis and HIV serology as well as vasculitis screen [3638]. An oral short trial of Levodopa will identify those individuals with dopa-responsive dystonia and neurophysiology can be useful if there is an associated neuropathy or suspicion of amyotrophic lateral sclerosis [39]. Table 3 summarises a list of conditions to be considered in the differential diagnosis and investigation of sporadic pure HSP.
Table 3
Conditions and investigations to be considered in the differential diagnosis of pure HSP
Structural and vascular
 Arterio-venous dural fistula [77]
MRI/angiogram
 Spinal or parasagittal tumour
MRI
 Spondylosis
MRI
Inflammatory
 Multiple Sclerosis
MRI, CSF
 Vasculitic Myelopathy [36]
Autoimmune profile
 Stiff person syndrome [78]
Neurophysiology, antibody testing
 Sarcoidosis [79]
MRI, CSF, chest X-ray
Metabolic (acquired and hereditary)
 Vitamin deficiency (B12, E)
Vitamin levels
 Nitrous oxide toxicity [80]
History, B12 level
 Adrenoleucodystrophy and other leucodystrophies [81, 82]
White cell enzymes, VLCFA, MRI
 Copper deficiency myelopathy [83]
Copper levels
Degenerative
 Primary lateral sclerosis
Neurophysiology and evolution of clinical picture
Infectious
 Tropical spastic paraparesis [37]
HTLV—1 serology, CSF
 HIV myelopathy [38]
HIV serology, CSF
 Syphilis
Syphilis serology, CSF
Other
 Radiation myelopathy [84]
History, imaging
 Spinocerebellar ataxias and other genetic conditions (see text and [39])
Genetic testing. Trial of l-dopa
For those individuals in whom the above baseline investigations are unremarkable and/or where the family history and clinical features point towards a specific genetic diagnosis, it is then appropriate to test for the suspected mutation.

Genetic investigations

There is considerable clinical utility for HSP families in detecting their causative mutation. It gives diagnostic certainty, prevents further unnecessary investigations and opens the possibility of predictive and prenatal testing. However, until very recently, in many healthcare systems, economic factors necessitated that genetic investigations were restricted to analysis of HSP genes in which there was the highest chance of detecting a mutation, as the expense of analysing rarely-causative genes was not justified by the low mutation detection rate per additional gene analysed. However, this has changed with the introduction of next-generation sequencing, which allows cost-effective analysis of many genes together, using approaches including (1) sequencing of large panels of genes, (2) sequencing of all coding exons in the genome, or (3) sequencing of entire genomes. At present, a common approach uses next-generation sequencing to screen exons of panels of genes causally associated with specific conditions or groups of conditions. These new approaches hold the prospect of substantially increasing the mutational pick-up rate for HSP patients. Once fully integrated into molecular diagnostic services, it is possible to envisage that genetic testing could be introduced at an early stage in patient work-up, perhaps avoiding the need for other costly or painful investigations if a definitive molecular genetic diagnosis is made.
Although these developments are undeniably positive, there are pitfalls and caveats associated with next-generation sequencing approaches. Sequence variation is very common in the genome (for example, many millions of single nucleotide variants have been identified) [40], with the vast majority of these sequence changes having a neutral effect with no clinical consequence. In addition, many of these sequence changes are individually rare. Thus, next-generation approaches typically detect numerous rare sequence variants; the more genes that are analysed, the larger this problem becomes. A major problem lies in deciding which, if any, of the detected sequence changes are pathogenic. In most cases, this can be resolved using bioinformatics and other approaches (e.g. identifying previously reported and validated mutations, excluding known rare population polymorphisms, determining the likely effect on the amino acid sequence of the encoded protein and how deleterious this effect is likely to be, excluding mutations that do not co-segregate with disease) [41]. However, inevitably in some cases, a single conclusively pathogenic mutation cannot be identified and one or more candidate pathogenic mutations remain. This is a particular issue with mutations of the missense class (in which only a single amino acid of the encoded protein is altered), since it can be difficult to predict whether the resulting effect on the protein will have functional consequences. Such sequence changes are often reported as variants of uncertain significance. Defining approaches to determining which of these mutations are pathogenic will be a major future challenge, as will be determining ethical approaches to whether and how the uncertainty associated with such sequence changes is reported back to patients.
In addition, exon-sequencing approaches do not detect all mutational classes. A particular problem arises in detecting large-scale deletions or duplications involving whole exons, or in detecting promoter or deep intronic mutations. This is an issue for at least some HSP genes, e.g. whole exon deletions are a common cause of SPG4/SPAST HSP (see above). We anticipate that in the relatively near future this issue will be resolved by whole-genome sequencing, which has the capacity to identify large deletions/duplications, as well as promoter and deep intronic mutations. In a research setting, genome sequencing may also allow identification of causative mutations in genes that have not previously been associated with the disease under consideration. In the meantime, additional testing for deletions/duplications should be considered in selected genes for appropriate patients, prioritised based on clinical features, in whom mutations have not been identified in exon-sequencing panels.
High-throughput sequencing approaches also may present ethical issues if so-called “incidental findings” (clinically relevant changes in genes unrelated to the condition being tested) are detected. These are particularly important when whole-exome or whole-genome approaches are taken, where, given sufficient numbers of tested subjects, it is inevitable that other potentially clinically significant abnormalities, e.g. mutations in cancer predisposing genes or detection of carrier status for autosomal recessive disease, will be detected. Whether and how such findings are reported back to patients requires careful consideration of their clinical validity and utility, and the ethical issues surrounding this are currently a topic of much debate [42].
Finally, while next-generation sequencing approaches have the potential to increase rates of molecular genetic diagnosis in HSP, they do not remove the need for careful phenotyping––this is still important, as it can help to focus bioinformatics analysis onto the most relevant genes. Careful phenotyping may also better define the clinical spectrum associated with pathogenic mutations in particular genes.

Rehabilitation and therapy for HSP patients

People with HSP complain of muscle stiffness, pain, spasms and cramps, tripping over their toes due to weakness of ankle dorsiflexion and hip flexion, loss of balance, effortful walking and progressively more flexed standing posture. Eventually, walking becomes impossible for some patients due to a combination of (a) spasticity, (b) weakness, particularly of ankle dorsiflexion, leading to (c) loss of range of movement at ankle, knee and hip, making it impossible to stand straight, and (d) loss of motor control leading to delayed postural reflexes and loss of balance.
A home exercise programme supervised by a physiotherapist, concentrating on stretches to maintain range of movement and reduce spasticity, accompanied by balance exercises in patients with more advanced disease, is the cornerstone of management. This is usually supported by a trial of oral muscle relaxants such as Baclofen, Tizanidine, or Gabapentin/Pregabalin. Problematic spasticity in specific groups of muscles, most commonly in the ankle plantarflexors and hip adductors, may be treated by Botulinum toxin without the risk of sedation associated with oral muscle relaxants [43]. The role of Botulinum toxin is to support and facilitate stretching and splinting, rather than to replace it. Functional electrical stimulation (FES) is popular among patients and physiotherapists and is as effective as simple off the shelf ankle foot orthoses (AFOs) in the early stages before calf shortening has developed: after that, FES is less useful as it cannot provide support or compensation during stance phase [4446].
Intrathecal Baclofen, delivered by an implanted pump, is the most effective method of reducing very high tone in the lower limbs, and can bring immediate relief of pain and improvement in sitting posture, along with a reduction in effort transferring from wheelchair to bed or car [47]. Effective control of muscle tone often improves quality of sleep for the patient and their partner, and permits stretching and splinting with the aim of preventing further deterioration of flexion contractors. The question of when to start intrathecal baclofen in this condition has not been addressed by published trials. While most often performed much later in the disease, a case can be made for implanting a pump relatively early to prevent the development of contractures, with a view to maintaining upright gait and retaining the option of supporting weak knee extensors and ankle dorsiflexors with orthoses [48].
Once ankle contractures have developed, and people can no longer stand with knees and hips straight while their heels are on the ground, heel wedging needs to be incorporated into shoes or AFOs to compensate. The lighter “off the shelf” AFOs, or FES of the peroneal nerve, are no longer appropriate in this situation as these only prevent passive plantarflexion during the swing phase of gait and are not designed to stabilise the foot and ankle during the stance phase of gait. The necessary custom moulded AFOs with compensatory heel wedging will often not fit in patients’ usual shoes; if larger trainers are impractical, expensive bespoke footwear may be required. Custom AFOs may also be designed to compensate for weak knee extensors using the ground reaction force to hold the lower leg in a more vertical position, but this is only possible if the knee still extends fully.

Conclusion

The last decade has seen astonishing progress in the identification of HSP genes. Coupled to the introduction of high-throughput sequencing approaches, we are quickly moving towards the ideal situation where every HSP patient will have a defined molecular diagnosis if they choose to have it. This will give important immediate benefits to HSP families, including diagnostic certainty, prevention of unnecessary additional investigations and accurate risk prediction for clinically unaffected family members. In the future, as HSP therapies emerge, it may also be a pre-requisite for the personalised selection of appropriate treatment. In the meantime, it is important that supportive therapy, which can make a real difference to patients’ lives, is not neglected.

Acknowledgments

We thank Rhys Roberts for reviewing the manuscript. This work was supported by grants from the UK Medical Research Council [MR/M00046X/1]; the Wellcome Trust [082381]; the Tom Wahlig Stiftung; and the UK HSP Support Group. The Cambridge Institute for Medical Research is supported by a Wellcome Trust Strategic Award [100140].

Conflicts of interest

The authors declare no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Neuer Inhalt

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
2.
Zurück zum Zitat Reid E, Rugarli E (2010) Hereditary spastic paraplegias. In: Valle D et al (eds) The online metabolic and molecular bases of inherited diseases. McGraw Hill, New York Reid E, Rugarli E (2010) Hereditary spastic paraplegias. In: Valle D et al (eds) The online metabolic and molecular bases of inherited diseases. McGraw Hill, New York
3.
Zurück zum Zitat Harding AE (1984) The hereditary ataxias and related disorders. Churchill Livingston, Edinburgh Harding AE (1984) The hereditary ataxias and related disorders. Churchill Livingston, Edinburgh
4.
Zurück zum Zitat Ruano L, Melo C, Silva MC, Coutinho P (2014) The global epidemiology of hereditary ataxia and spastic paraplegia: a systematic review of prevalence studies. Neuroepidemiology 42:174–183CrossRefPubMed Ruano L, Melo C, Silva MC, Coutinho P (2014) The global epidemiology of hereditary ataxia and spastic paraplegia: a systematic review of prevalence studies. Neuroepidemiology 42:174–183CrossRefPubMed
5.
Zurück zum Zitat Harding AE (1981) Hereditary “pure” spastic paraplegia: a clinical and genetic study of 22 families. J Neurol Neurosurg Psychiatry 44:871–883PubMedCentralCrossRefPubMed Harding AE (1981) Hereditary “pure” spastic paraplegia: a clinical and genetic study of 22 families. J Neurol Neurosurg Psychiatry 44:871–883PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Reid E, Grayson C, Rogers MT, Rubinsztein DC (1999) Locus-phenotype correlations in autosomal dominant pure hereditary spastic paraplegia. A clinical and molecular genetic study of 28 United Kingdom families. Brain 122(Pt 9):1741–1755CrossRefPubMed Reid E, Grayson C, Rogers MT, Rubinsztein DC (1999) Locus-phenotype correlations in autosomal dominant pure hereditary spastic paraplegia. A clinical and molecular genetic study of 28 United Kingdom families. Brain 122(Pt 9):1741–1755CrossRefPubMed
7.
Zurück zum Zitat Durr A et al (1994) The phenotype of “pure” autosomal dominant spastic paraplegia. Neurology 44:1274–1277CrossRefPubMed Durr A et al (1994) The phenotype of “pure” autosomal dominant spastic paraplegia. Neurology 44:1274–1277CrossRefPubMed
9.
Zurück zum Zitat Fink JK et al (1996) Hereditary spastic paraplegia: advances in genetic research. Hereditary Spastic Paraplegia Working group. Neurology 46:1507–1514CrossRefPubMed Fink JK et al (1996) Hereditary spastic paraplegia: advances in genetic research. Hereditary Spastic Paraplegia Working group. Neurology 46:1507–1514CrossRefPubMed
10.
Zurück zum Zitat Boukhris A et al (2009) Tunisian hereditary spastic paraplegias: clinical variability supported by genetic heterogeneity. Clin Genet 75:527–536CrossRefPubMed Boukhris A et al (2009) Tunisian hereditary spastic paraplegias: clinical variability supported by genetic heterogeneity. Clin Genet 75:527–536CrossRefPubMed
12.
Zurück zum Zitat Nielsen JE et al (2004) Hereditary spastic paraplegia with cerebellar ataxia: a complex phenotype associated with a new SPG4 gene mutation. Eur J Neurol 11:817–824CrossRefPubMed Nielsen JE et al (2004) Hereditary spastic paraplegia with cerebellar ataxia: a complex phenotype associated with a new SPG4 gene mutation. Eur J Neurol 11:817–824CrossRefPubMed
13.
Zurück zum Zitat Ivanova N et al (2007) Hereditary spastic paraplegia 3A associated with axonal neuropathy. Arch Neurol 64:706–713CrossRefPubMed Ivanova N et al (2007) Hereditary spastic paraplegia 3A associated with axonal neuropathy. Arch Neurol 64:706–713CrossRefPubMed
14.
Zurück zum Zitat Goizet C et al (2011) REEP1 mutations in SPG31: frequency, mutational spectrum, and potential association with mitochondrial morpho-functional dysfunction. Hum Mutat 32:1118–1127CrossRefPubMed Goizet C et al (2011) REEP1 mutations in SPG31: frequency, mutational spectrum, and potential association with mitochondrial morpho-functional dysfunction. Hum Mutat 32:1118–1127CrossRefPubMed
15.
Zurück zum Zitat Erichsen AK, Koht J, Stray-Pedersen A, Abdelnoor M, Tallaksen CM (2009) Prevalence of hereditary ataxia and spastic paraplegia in southeast Norway: a population-based study. Brain 132:1577–1588CrossRefPubMed Erichsen AK, Koht J, Stray-Pedersen A, Abdelnoor M, Tallaksen CM (2009) Prevalence of hereditary ataxia and spastic paraplegia in southeast Norway: a population-based study. Brain 132:1577–1588CrossRefPubMed
16.
Zurück zum Zitat Depienne C et al (2007) A de novo SPAST mutation leading to somatic mosaicism is associated with a later age at onset in HSP. Neurogenetics 8:231–233CrossRefPubMed Depienne C et al (2007) A de novo SPAST mutation leading to somatic mosaicism is associated with a later age at onset in HSP. Neurogenetics 8:231–233CrossRefPubMed
18.
Zurück zum Zitat Beetz C et al (2006) High frequency of partial SPAST deletions in autosomal dominant hereditary spastic paraplegia. Neurology 67:1926–1930CrossRefPubMed Beetz C et al (2006) High frequency of partial SPAST deletions in autosomal dominant hereditary spastic paraplegia. Neurology 67:1926–1930CrossRefPubMed
19.
Zurück zum Zitat Sulek A et al (2013) Screening for the hereditary spastic paraplaegias SPG4 and SPG3A with the multiplex ligation-dependent probe amplification technique in a large population of affected individuals. Neurol Sci 34:239–242CrossRefPubMed Sulek A et al (2013) Screening for the hereditary spastic paraplaegias SPG4 and SPG3A with the multiplex ligation-dependent probe amplification technique in a large population of affected individuals. Neurol Sci 34:239–242CrossRefPubMed
20.
Zurück zum Zitat Stevanin G et al (2007) Mutations in SPG11, encoding spatacsin, are a major cause of spastic paraplegia with thin corpus callosum. Nat Genet 39:366–372CrossRefPubMed Stevanin G et al (2007) Mutations in SPG11, encoding spatacsin, are a major cause of spastic paraplegia with thin corpus callosum. Nat Genet 39:366–372CrossRefPubMed
21.
Zurück zum Zitat Stevanin G et al (2008) Mutations in SPG11 are frequent in autosomal recessive spastic paraplegia with thin corpus callosum, cognitive decline and lower motor neuron degeneration. Brain 131:772–784CrossRefPubMed Stevanin G et al (2008) Mutations in SPG11 are frequent in autosomal recessive spastic paraplegia with thin corpus callosum, cognitive decline and lower motor neuron degeneration. Brain 131:772–784CrossRefPubMed
22.
Zurück zum Zitat Hanein S et al (2008) Identification of the SPG15 gene, encoding spastizin, as a frequent cause of complicated autosomal-recessive spastic paraplegia, including Kjellin syndrome. Am J Hum Genet 82:992–1002PubMedCentralCrossRefPubMed Hanein S et al (2008) Identification of the SPG15 gene, encoding spastizin, as a frequent cause of complicated autosomal-recessive spastic paraplegia, including Kjellin syndrome. Am J Hum Genet 82:992–1002PubMedCentralCrossRefPubMed
23.
25.
Zurück zum Zitat Pensato V et al (2014) Overlapping phenotypes in complex spastic paraplegias SPG11, SPG15, SPG35 and SPG48. Brain 137:1907–1920CrossRefPubMed Pensato V et al (2014) Overlapping phenotypes in complex spastic paraplegias SPG11, SPG15, SPG35 and SPG48. Brain 137:1907–1920CrossRefPubMed
26.
Zurück zum Zitat van de Warrenburg BPC et al (2014) EFNS/ENS Consensus on the diagnosis and management of chronic ataxias in adulthood. Eur J Neurol 21:552–562CrossRefPubMed van de Warrenburg BPC et al (2014) EFNS/ENS Consensus on the diagnosis and management of chronic ataxias in adulthood. Eur J Neurol 21:552–562CrossRefPubMed
27.
Zurück zum Zitat Sedel F, Lyon-Caen O, Saudubray JM (2007) Therapy insight: inborn errors of metabolism in adult neurology––a clinical approach focused on treatable diseases. Nature clinical practice. Neurology 3:279–290PubMed Sedel F, Lyon-Caen O, Saudubray JM (2007) Therapy insight: inborn errors of metabolism in adult neurology––a clinical approach focused on treatable diseases. Nature clinical practice. Neurology 3:279–290PubMed
28.
Zurück zum Zitat de Bot ST, van de Warrenburg BP, Kremer HP, Willemsen MA (2010) Child neurology: hereditary spastic paraplegia in children. Neurology 75:e75–e79CrossRefPubMed de Bot ST, van de Warrenburg BP, Kremer HP, Willemsen MA (2010) Child neurology: hereditary spastic paraplegia in children. Neurology 75:e75–e79CrossRefPubMed
29.
Zurück zum Zitat Shimazaki H et al (2014) Autosomal-recessive complicated spastic paraplegia with a novel lysosomal trafficking regulator gene mutation. J Neurol Neurosurg Psychiatry 85:1024–1028CrossRefPubMed Shimazaki H et al (2014) Autosomal-recessive complicated spastic paraplegia with a novel lysosomal trafficking regulator gene mutation. J Neurol Neurosurg Psychiatry 85:1024–1028CrossRefPubMed
30.
Zurück zum Zitat Goizet C et al (2009) CYP7B1 mutations in pure and complex forms of hereditary spastic paraplegia type 5. Brain 132:1589–1600CrossRefPubMed Goizet C et al (2009) CYP7B1 mutations in pure and complex forms of hereditary spastic paraplegia type 5. Brain 132:1589–1600CrossRefPubMed
31.
Zurück zum Zitat Wang Y-G et al (2009) Six cases of SCA3/MJD patients that mimic hereditary spastic paraplegia in clinic. J Neurol Sci 285:121–124CrossRefPubMed Wang Y-G et al (2009) Six cases of SCA3/MJD patients that mimic hereditary spastic paraplegia in clinic. J Neurol Sci 285:121–124CrossRefPubMed
32.
Zurück zum Zitat de Bot ST, Willemsen MA, Vermeer S, Kremer HP, van de Warrenburg BP (2012) Reviewing the genetic causes of spastic-ataxias. Neurology 79:1507–1514CrossRefPubMed de Bot ST, Willemsen MA, Vermeer S, Kremer HP, van de Warrenburg BP (2012) Reviewing the genetic causes of spastic-ataxias. Neurology 79:1507–1514CrossRefPubMed
33.
Zurück zum Zitat Casari G et al (1998) Spastic paraplegia and OXPHOS impairment caused by mutations in paraplegin, a nuclear-encoded mitochondrial metalloprotease. Cell 93:973–983CrossRefPubMed Casari G et al (1998) Spastic paraplegia and OXPHOS impairment caused by mutations in paraplegin, a nuclear-encoded mitochondrial metalloprotease. Cell 93:973–983CrossRefPubMed
34.
Zurück zum Zitat Wilkinson PA et al (2004) A clinical, genetic and biochemical study of SPG7 mutations in hereditary spastic paraplegia. Brain 127:973–980CrossRefPubMed Wilkinson PA et al (2004) A clinical, genetic and biochemical study of SPG7 mutations in hereditary spastic paraplegia. Brain 127:973–980CrossRefPubMed
35.
36.
Zurück zum Zitat Ropper AH, Ayata C, Adelman L (2003) Vasculitis of the spinal cord. Arch Neurol 60:1791–1794CrossRefPubMed Ropper AH, Ayata C, Adelman L (2003) Vasculitis of the spinal cord. Arch Neurol 60:1791–1794CrossRefPubMed
37.
38.
39.
Zurück zum Zitat Furukawa Y, Graf WD, Wong H, Shimadzu M, Kish SJ (2001) Dopa-responsive dystonia simulating spastic paraplegia due to tyrosine hydroxylase (TH) gene mutations. Neurology 56:260–263CrossRefPubMed Furukawa Y, Graf WD, Wong H, Shimadzu M, Kish SJ (2001) Dopa-responsive dystonia simulating spastic paraplegia due to tyrosine hydroxylase (TH) gene mutations. Neurology 56:260–263CrossRefPubMed
40.
Zurück zum Zitat Liu X, Jian X, Boerwinkle E (2013) dbNSFP v2.0: a database of human non-synonymous SNVs and their functional predictions and annotations. Hum Mutat 34:E2393–E2402PubMedCentralCrossRefPubMed Liu X, Jian X, Boerwinkle E (2013) dbNSFP v2.0: a database of human non-synonymous SNVs and their functional predictions and annotations. Hum Mutat 34:E2393–E2402PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Berg JS et al (2013) Processes and preliminary outputs for identification of actionable genes as incidental findings in genomic sequence data in the Clinical Sequencing Exploratory Research Consortium. Genet Med 15:860–867PubMedCentralCrossRefPubMed Berg JS et al (2013) Processes and preliminary outputs for identification of actionable genes as incidental findings in genomic sequence data in the Clinical Sequencing Exploratory Research Consortium. Genet Med 15:860–867PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Geva-Dayan K, Domenievitz D, Zahalka R, Fattal-Valevski A (2010) Botulinum toxin injections for pediatric patients with hereditary spastic paraparesis. J Child Neurol 25:969–975CrossRefPubMed Geva-Dayan K, Domenievitz D, Zahalka R, Fattal-Valevski A (2010) Botulinum toxin injections for pediatric patients with hereditary spastic paraparesis. J Child Neurol 25:969–975CrossRefPubMed
44.
Zurück zum Zitat Kluding PM et al (2013) Foot drop stimulation versus ankle foot orthosis after stroke: 30-week outcomes. Stroke 44:1660–1669CrossRefPubMed Kluding PM et al (2013) Foot drop stimulation versus ankle foot orthosis after stroke: 30-week outcomes. Stroke 44:1660–1669CrossRefPubMed
45.
Zurück zum Zitat Karimi MT (2013) Functional walking ability of paraplegic patients: comparison of functional electrical stimulation versus mechanical orthoses. Eur J Orthop Surg Traumatol orthop traumatol 23:631–638CrossRef Karimi MT (2013) Functional walking ability of paraplegic patients: comparison of functional electrical stimulation versus mechanical orthoses. Eur J Orthop Surg Traumatol orthop traumatol 23:631–638CrossRef
46.
Zurück zum Zitat Bethoux F et al (2014) The effects of peroneal nerve functional electrical stimulation versus ankle-foot orthosis in patients with chronic stroke: a randomized controlled trial. Neurorehab Neural Repair 28:688–697CrossRef Bethoux F et al (2014) The effects of peroneal nerve functional electrical stimulation versus ankle-foot orthosis in patients with chronic stroke: a randomized controlled trial. Neurorehab Neural Repair 28:688–697CrossRef
47.
Zurück zum Zitat Meythaler JM et al (1992) Intrathecal baclofen in hereditary spastic paraparesis. Arch Phys Med Rehabil 73:794–797PubMed Meythaler JM et al (1992) Intrathecal baclofen in hereditary spastic paraparesis. Arch Phys Med Rehabil 73:794–797PubMed
48.
Zurück zum Zitat Lambrecq V et al. (2007) Intrathecal baclofen in hereditary spastic paraparesis: benefits and limitations. Annales de readaptation et de medecine physique : revue scientifique de la Societe francaise de reeducation fonctionnelle de readaptation et de medecine physique 50:577–581 Lambrecq V et al. (2007) Intrathecal baclofen in hereditary spastic paraparesis: benefits and limitations. Annales de readaptation et de medecine physique : revue scientifique de la Societe francaise de reeducation fonctionnelle de readaptation et de medecine physique 50:577–581
49.
Zurück zum Zitat Fonknechten N et al (2000) Spectrum of SPG4 mutations in autosomal dominant spastic paraplegia. Hum Mol Genet 9:637–644CrossRefPubMed Fonknechten N et al (2000) Spectrum of SPG4 mutations in autosomal dominant spastic paraplegia. Hum Mol Genet 9:637–644CrossRefPubMed
50.
Zurück zum Zitat Patrono C et al (2005) Autosomal dominant hereditary spastic paraplegia: DHPLC-based mutation analysis of SPG4 reveals eleven novel mutations. Hum Mutat 25:506CrossRefPubMed Patrono C et al (2005) Autosomal dominant hereditary spastic paraplegia: DHPLC-based mutation analysis of SPG4 reveals eleven novel mutations. Hum Mutat 25:506CrossRefPubMed
51.
Zurück zum Zitat Sauter S et al (2002) Mutation analysis of the spastin gene (SPG4) in patients in Germany with autosomal dominant hereditary spastic paraplegia. Hum Mutat 20:127–132CrossRefPubMed Sauter S et al (2002) Mutation analysis of the spastin gene (SPG4) in patients in Germany with autosomal dominant hereditary spastic paraplegia. Hum Mutat 20:127–132CrossRefPubMed
52.
Zurück zum Zitat Shoukier M et al (2009) Expansion of mutation spectrum, determination of mutation cluster regions and predictive structural classification of SPAST mutations in hereditary spastic paraplegia. Eur J Hum Genet 17:187–194PubMedCentralCrossRefPubMed Shoukier M et al (2009) Expansion of mutation spectrum, determination of mutation cluster regions and predictive structural classification of SPAST mutations in hereditary spastic paraplegia. Eur J Hum Genet 17:187–194PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Meijer HA, Thomas AA (2002) Control of eukaryotic protein synthesis by upstream open reading frames in the 5′-untranslated region of an mRNA. Biochem J367:1–11CrossRef Meijer HA, Thomas AA (2002) Control of eukaryotic protein synthesis by upstream open reading frames in the 5′-untranslated region of an mRNA. Biochem J367:1–11CrossRef
54.
Zurück zum Zitat Park SY et al (2005) Mutation analysis of SPG4 and SPG3A genes and its implication in molecular diagnosis of Korean patients with hereditary spastic paraplegia. Arch Neurol 62:1118–1121CrossRefPubMed Park SY et al (2005) Mutation analysis of SPG4 and SPG3A genes and its implication in molecular diagnosis of Korean patients with hereditary spastic paraplegia. Arch Neurol 62:1118–1121CrossRefPubMed
55.
Zurück zum Zitat Depienne C et al (2006) Spastin mutations are frequent in sporadic spastic paraparesis and their spectrum is different from that observed in familial cases. J Med Genet 43:259–265PubMedCentralCrossRefPubMed Depienne C et al (2006) Spastin mutations are frequent in sporadic spastic paraparesis and their spectrum is different from that observed in familial cases. J Med Genet 43:259–265PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Crippa F et al (2006) Eight novel mutations in SPG4 in a large sample of patients with hereditary spastic paraplegia. Arch Neurol 63:750–755CrossRefPubMed Crippa F et al (2006) Eight novel mutations in SPG4 in a large sample of patients with hereditary spastic paraplegia. Arch Neurol 63:750–755CrossRefPubMed
57.
Zurück zum Zitat Brugman F et al (2005) Spastin mutations in sporadic adult-onset upper motor neuron syndromes. Ann Neurol 58:865–869CrossRefPubMed Brugman F et al (2005) Spastin mutations in sporadic adult-onset upper motor neuron syndromes. Ann Neurol 58:865–869CrossRefPubMed
58.
Zurück zum Zitat Braschinsky M, Luus SM, Gross-Paju K, Haldre S (2009) The prevalence of hereditary spastic paraplegia and the occurrence of SPG4 mutations in Estonia. Neuroepidemiology 32:89–93CrossRefPubMed Braschinsky M, Luus SM, Gross-Paju K, Haldre S (2009) The prevalence of hereditary spastic paraplegia and the occurrence of SPG4 mutations in Estonia. Neuroepidemiology 32:89–93CrossRefPubMed
59.
Zurück zum Zitat Abel A et al (2004) Early onset autosomal dominant spastic paraplegia caused by novel mutations in SPG3A. Neurogenetics 5:239–243CrossRefPubMed Abel A et al (2004) Early onset autosomal dominant spastic paraplegia caused by novel mutations in SPG3A. Neurogenetics 5:239–243CrossRefPubMed
60.
Zurück zum Zitat Durr A et al (2004) Atlastin1 mutations are frequent in young-onset autosomal dominant spastic paraplegia. Arch Neurol 61:1867–1872CrossRefPubMed Durr A et al (2004) Atlastin1 mutations are frequent in young-onset autosomal dominant spastic paraplegia. Arch Neurol 61:1867–1872CrossRefPubMed
61.
Zurück zum Zitat Zhao X et al (2001) Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat Genet 29:326–331CrossRefPubMed Zhao X et al (2001) Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat Genet 29:326–331CrossRefPubMed
62.
Zurück zum Zitat Schlang KJ, Arning L, Epplen JT, Stemmler S (2008) Autosomal dominant hereditary spastic paraplegia: novel mutations in the REEP1 gene (SPG31). BMC Med Genet 9:71PubMedCentralCrossRefPubMed Schlang KJ, Arning L, Epplen JT, Stemmler S (2008) Autosomal dominant hereditary spastic paraplegia: novel mutations in the REEP1 gene (SPG31). BMC Med Genet 9:71PubMedCentralCrossRefPubMed
63.
64.
Zurück zum Zitat Hewamadduma C et al (2009) New pedigrees and novel mutation expand the phenotype of REEP1-associated hereditary spastic paraplegia (HSP). Neurogenetics 10:105–110CrossRefPubMed Hewamadduma C et al (2009) New pedigrees and novel mutation expand the phenotype of REEP1-associated hereditary spastic paraplegia (HSP). Neurogenetics 10:105–110CrossRefPubMed
65.
Zurück zum Zitat Zuchner S et al (2006) Mutations in the novel mitochondrial protein REEP1 cause hereditary spastic paraplegia type 31. Am J Hum Genet 79:365–369PubMedCentralCrossRefPubMed Zuchner S et al (2006) Mutations in the novel mitochondrial protein REEP1 cause hereditary spastic paraplegia type 31. Am J Hum Genet 79:365–369PubMedCentralCrossRefPubMed
66.
Zurück zum Zitat Schule R et al (2008) SPG10 is a rare cause of spastic paraplegia in European families. J Neurol Neurosurg Psychiatry 79:584–587CrossRefPubMed Schule R et al (2008) SPG10 is a rare cause of spastic paraplegia in European families. J Neurol Neurosurg Psychiatry 79:584–587CrossRefPubMed
67.
Zurück zum Zitat Tessa A et al (2008) A novel KIF5A/SPG10 mutation in spastic paraplegia associated with axonal neuropathy. J Neurol 255:1090–1092CrossRefPubMed Tessa A et al (2008) A novel KIF5A/SPG10 mutation in spastic paraplegia associated with axonal neuropathy. J Neurol 255:1090–1092CrossRefPubMed
68.
Zurück zum Zitat Goizet C et al (2009) Complicated forms of autosomal dominant hereditary spastic paraplegia are frequent in SPG10. Hum Mutat 30:E376–E385CrossRefPubMed Goizet C et al (2009) Complicated forms of autosomal dominant hereditary spastic paraplegia are frequent in SPG10. Hum Mutat 30:E376–E385CrossRefPubMed
69.
70.
Zurück zum Zitat de Bot S et al (2013) Pure adult-onset Spastic Paraplegia caused by a novel mutation in the KIAA0196 (SPG8) gene. J Neurol 260:1765–1769CrossRefPubMed de Bot S et al (2013) Pure adult-onset Spastic Paraplegia caused by a novel mutation in the KIAA0196 (SPG8) gene. J Neurol 260:1765–1769CrossRefPubMed
71.
Zurück zum Zitat Montenegro G et al (2012) Mutations in the ER-shaping protein reticulon 2 cause the axon-degenerative disorder hereditary spastic paraplegia type 12. J Clin Investig 122:538–544PubMedCentralCrossRefPubMed Montenegro G et al (2012) Mutations in the ER-shaping protein reticulon 2 cause the axon-degenerative disorder hereditary spastic paraplegia type 12. J Clin Investig 122:538–544PubMedCentralCrossRefPubMed
72.
Zurück zum Zitat Beetz C et al (2008) Screening of hereditary spastic paraplegia patients for alterations at NIPA1 mutational hotspots. J Neurol Sci 268:131–135CrossRefPubMed Beetz C et al (2008) Screening of hereditary spastic paraplegia patients for alterations at NIPA1 mutational hotspots. J Neurol Sci 268:131–135CrossRefPubMed
73.
Zurück zum Zitat Klebe S et al (2007) NIPA1 (SPG6) mutations are a rare cause of autosomal dominant spastic paraplegia in Europe. Neurogenetics 8:155–157CrossRefPubMed Klebe S et al (2007) NIPA1 (SPG6) mutations are a rare cause of autosomal dominant spastic paraplegia in Europe. Neurogenetics 8:155–157CrossRefPubMed
74.
Zurück zum Zitat Brugman F et al (2008) Paraplegin mutations in sporadic adult-onset upper motor neuron syndromes. Neurology 71:1500–1505CrossRefPubMed Brugman F et al (2008) Paraplegin mutations in sporadic adult-onset upper motor neuron syndromes. Neurology 71:1500–1505CrossRefPubMed
75.
Zurück zum Zitat Arnoldi A et al (2008) A clinical, genetic, and biochemical characterization of SPG7 mutations in a large cohort of patients with hereditary spastic paraplegia. Hum Mutat 29:522–531CrossRefPubMed Arnoldi A et al (2008) A clinical, genetic, and biochemical characterization of SPG7 mutations in a large cohort of patients with hereditary spastic paraplegia. Hum Mutat 29:522–531CrossRefPubMed
76.
Zurück zum Zitat Elleuch N et al (2006) Mutation analysis of the paraplegin gene (SPG7) in patients with hereditary spastic paraplegia. Neurology 66:654–659CrossRefPubMed Elleuch N et al (2006) Mutation analysis of the paraplegin gene (SPG7) in patients with hereditary spastic paraplegia. Neurology 66:654–659CrossRefPubMed
77.
Zurück zum Zitat Caragine LP Jr, Halbach VV, Ng PP, Dowd CF (2002) Vascular myelopathies-vascular malformations of the spinal cord: presentation and endovascular surgical management. Semin Neurol 22:123–132CrossRefPubMed Caragine LP Jr, Halbach VV, Ng PP, Dowd CF (2002) Vascular myelopathies-vascular malformations of the spinal cord: presentation and endovascular surgical management. Semin Neurol 22:123–132CrossRefPubMed
78.
Zurück zum Zitat Barker RA, Revesz T, Thom M, Marsden CD, Brown P (1998) Review of 23 patients affected by the stiff man syndrome: clinical subdivision into stiff trunk (man) syndrome, stiff limb syndrome, and progressive encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 65:633–640PubMedCentralCrossRefPubMed Barker RA, Revesz T, Thom M, Marsden CD, Brown P (1998) Review of 23 patients affected by the stiff man syndrome: clinical subdivision into stiff trunk (man) syndrome, stiff limb syndrome, and progressive encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 65:633–640PubMedCentralCrossRefPubMed
79.
Zurück zum Zitat Saleh S, Saw C, Marzouk K, Sharma O (2006) Sarcoidosis of the spinal cord: literature review and report of eight cases. J Natl Med Assoc 98:965–976PubMedCentralPubMed Saleh S, Saw C, Marzouk K, Sharma O (2006) Sarcoidosis of the spinal cord: literature review and report of eight cases. J Natl Med Assoc 98:965–976PubMedCentralPubMed
80.
Zurück zum Zitat Pema PJ, Horak HA, Wyatt RH (1998) Myelopathy caused by nitrous oxide toxicity. AJNR Am J Neuroradiol 19:894–896PubMed Pema PJ, Horak HA, Wyatt RH (1998) Myelopathy caused by nitrous oxide toxicity. AJNR Am J Neuroradiol 19:894–896PubMed
81.
Zurück zum Zitat Shaw-Smith CJ, Lewis SJ, Reid E (2004) X-linked adrenoleukodystrophy presenting as autosomal dominant pure hereditary spastic paraparesis. J Neurol Neurosurg Psychiatry 75:686–688PubMedCentralCrossRefPubMed Shaw-Smith CJ, Lewis SJ, Reid E (2004) X-linked adrenoleukodystrophy presenting as autosomal dominant pure hereditary spastic paraparesis. J Neurol Neurosurg Psychiatry 75:686–688PubMedCentralCrossRefPubMed
82.
Zurück zum Zitat Müller vom Hagen J, Karle KN, Schüle R, Krägeloh-Mann I, Schöls L (2014) Leukodystrophies underlying cryptic spastic paraparesis: frequency and phenotype in 76 patients. Eur J Neurol 21:983–988CrossRefPubMed Müller vom Hagen J, Karle KN, Schüle R, Krägeloh-Mann I, Schöls L (2014) Leukodystrophies underlying cryptic spastic paraparesis: frequency and phenotype in 76 patients. Eur J Neurol 21:983–988CrossRefPubMed
Metadaten
Titel
Diagnosis, investigation and management of hereditary spastic paraplegias in the era of next-generation sequencing
verfasst von
Anke Hensiek
Stephen Kirker
Evan Reid
Publikationsdatum
01.07.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Neurology / Ausgabe 7/2015
Print ISSN: 0340-5354
Elektronische ISSN: 1432-1459
DOI
https://doi.org/10.1007/s00415-014-7598-y

Weitere Artikel der Ausgabe 7/2015

Journal of Neurology 7/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.