You are on page 1of 7

J Neurol (2013) 260:26842690 DOI 10.

1007/s00415-013-7113-x

MEDICAL PROGRESS IN THE JOURNAL OF NEUROLOGY

Inherited neuropathies: an update


Anna Sagnelli Giuseppe Piscosquito Davide Pareyson

Received: 5 September 2013 / Revised: 11 September 2013 / Accepted: 12 September 2013 / Published online: 24 September 2013 Springer-Verlag Berlin Heidelberg 2013

Abstract In this review, progress in hereditary neuropathy research published in the Journal of Neurology over the last 18 months is summarised. Keywords Charcot-Marie-Tooth disease Familial amyloid polyneuropathy Mitochondrial disease Tangier disease Allgrove syndrome ARSACS

Introduction The ability to diagnose and better understand the pathophysiology of hereditary neuropathies has greatly increased over the last few years. This article reviews and summarises new ndings in the eld with particular reference to papers published in the Journal of Neurology over the last 18 months.

Charcot-Marie-Tooth disease and related neuropathies Charcot-Marie-Tooth disease (CMT) and cognate disorders are the most prevalent hereditary neuropathies, affecting one individual in every 2,500. Almost 70 genes have been associated with this heterogeneous group of neuropathies, and many of them have been discovered during the last 34 years, thanks to next generation sequencing technology such as whole exome sequencing [1, 2].

A. Sagnelli G. Piscosquito D. Pareyson (&) Clinic of Central and Peripheral Degenerative Neuropathies Unit, Department of Clinical Neurosciences, IRCCS Foundation, C. Besta Neurological Institute, via Celoria 11, 20133 Milan, Italy e-mail: davide.pareyson@istituto-besta.it

In an original and practical approach, Fischer et al. [3] exploited and adapted single-nucleotide polymorphism (SNP) array-based whole genome homozygosity mapping as a rst screening step in the investigation of a series of 24 CMT patients with sporadic or recessive presentation, and could detect a genetic mutation in CMT-related genes in 63 % of cases. In their series, the demyelinating recessive CMT4C associated with mutations in the SH3TC2 gene was the most common genetic subtype, followed by CMT4A related to GDAP1 mutations. They conrmed that HSPB1 mutations, associated with a dominant distal hereditary motor neuropathy (dHMN) or axonal CMT (CMT2), may be rarely associated with recessive CMT2/ dHMN. They also found another instance of the rare recessive CMT related to Mitofusin 2 (MFN2) mutations, which usually cause dominantly inherited neuropathies. Davidson and colleagues studied a very large series of 140 index patients with Hereditary Sensory and Autonomic Neuropathy (HSAN), a heterogeneous group of rare disorders characterised by pure or predominant sensory involvement, with at least ve different forms recognised [4]. The authors investigated by direct sequencing the frequency distribution of six HSAN genes: SPTCL1 associated with HSAN IA, RAB7 (CMT2B), WNK1/HSN2 (HSAN IIA), FAM134B (HSAN IIB), NTRK1 (HSAN IV or Congenital insensitivity to pain with anhidrosis), and NFGB (HSAN V). Two other HSAN genes were not investigated, as there was no indication to do this from their clinical presentation. The authors found a pathogenic mutation in at least 14 % of the cases (a rate similar to the 19 % of the previous series by Rotthier and coauthors [5]), a still low yield that indicates that further HSAN genes are to be identied, and indeed ve genes have been recently uncovered after study completion [6]. SPTLC1 was the most commonly mutated gene in UK patients (13 index

123

J Neurol (2013) 260:26842690

2685

cases) and the p.Cys133Trp founder mutation was the most prevalent. NTRK1 was the next commonly mutated gene (6 %), predominantly in patients of Saudi Arabian descent, while mutations in all the other genes were rare, accounting for \ 2 % each. Only one patient had a mutation in RAB7, which conversely was the most common mutated gene in the series of Rotthier et al. [5], partly due to a founder mutation in Austria. Two patients had novel recessive mutations in WNK1/HSN2, associated with the rare earlyonset recessive HSAN type IIA form, characterised by sensory loss to all modalities leading to ulcers, acromutilation and Charcot joints [7, 8]. HSAN IIA was initially thought to be related to mutations in the HSN2 gene, but it later became evident that, as Shekarabi and colleagues point out [9], HSN2 is only a coding exon of the larger Protein Kinase Lysine-Decient 1 (WNK1) gene. The WNK1/HSN2 isoform is specic to the nervous system and the alternatively spliced HSN2 exon is expressed only in neurons [6]. Genetic testing has become important in everyday clinical practice and it is relevant even for late-onset neuropathies, as a genetic cause is recognised in an increasing number of such cases. This is well shown by the paper by Marttila and colleagues, who report one Finnish family and one German patient with late-onset predominantly axonal CMT associated with the same novel MPZ mutation (p.Arg106Cys) [10]. MPZ mutations are commonly associated with early-onset demyelinating or hypomyelinating CMT1B, but they also lead to CMT2 of much later onset [1, 11]. In this report, age of onset ranged between 48 and 67 years; two patients had sensorineural hearing loss, possibly related to CMT. The genetic nature of the neuropathy is not obvious in sporadic cases and MPZ mutations need to be considered in the differential diagnosis of late-onset chronic axonal neuropathies [12]. The relationship between inherited and dysimmune neuropathies is matter of investigation and debate. There are some reports suggesting possible superimposition of dysimmune neuropathies, such as chronic inammatory demyelinating polyradiculoneuropathy (CIDP), on different CMT types [13, 14]. The X-linked CMT type, CMTX, is particularly liable to be misdiagnosed with CIDP, because it is characterised by non-homogeneous conduction slowing, sometimes accompanied by temporal dispersion and conduction blocks [1, 15]. Miki and colleagues reported a CMTX family carrying the novel p.Leu76Arg GJB1 mutation in which two hemizygous brothers had rather sudden progression of decits in their upper limbs and seemed to improve after repeat intravenous administration of IgG (IVIg), at least on clinical grounds, as no electrophysiological follow-up was performed [16]; the mutation-carrier sister was also treated with IVIg, but only minimal, if any, improvement was observed. Sudden

worsening occurs in CMT, and evidence of inammation is sometimes found at nerve biopsy; it is absolutely unclear and far from established whether this really represents an immune-mediated phenomenon requiring immunomodulation. Therefore, caution is to be recommended in interpreting CMT uctuations, and treatment with IVIg or immunosuppression must be avoided unless there are sure reasons for expecting denite and lasting improvement, and any claimed efcacy must be clearly objectively documented. Among the techniques that can be helpful in distinguishing CMT from CIDP, ultrasonography of nerves and roots might have a role. Sugimoto and colleagues employed median and ulnar nerve and cervical root ultrasonography in ten demyelinating CMT and 16 CIDP patients in an attempt to differentiate the two diseases. Demyelinating CMT patients showed signicant larger nerve size at all levels in median and ulnar nerves as compared to CIDP patients [17]. An interesting case of co-occurrence of CMTX and multiple sclerosis (MS) was described by Weishaupt and colleagues [18]; their case is the third instance of MS in CMTX patients and the authors discussed whether this is simply coincidental or if there is a predisposing role of abnormal GJB1 in oligodendrocytes. Notably, dramatic central nervous system dysfunction may rarely occur in CMTX males, but is transient and accompanying magnetic resonance imaging (MRI) abnormalities are widespread and usually resolve after clinical regression [19]. In this patient, however, MRI and cerebrospinal uid (CSF) abnormalities were typical of MS, so that treatment with natalizumab was started and appeared to be successful.

Schwannomas and inherited neuropathies Improvement in techniques of neuroimaging for peripheral nerves is allowing for detailed morphological studies, and may be of help in clarifying the relationship between tumours of nerves and hereditary neuropathies. It is of interest that there are occasional reports of isolated or multiple schwannomas occurring in patients affected by hereditary neuropathies of different nature, i.e. CMT type 1A associated with the PMP22 duplication [20], Hereditary Neuropathy with liability to Pressure Palsies (HNPP) caused by the reciprocal PMP22 deletion [21], and CMT related to INF2 (Inverted Forming 2) mutations [22]. Recently, Ohyagi and colleagues [23] described an HNPP patient who had two suspected schwannomas close to entrapment sites, namely the peroneal nerve below the bular head and the radial nerve at the elbow. The authors hypothesised that up-regulation and down-regulation of PMP22, which is also involved in cell cycle regulation,

123

2686

J Neurol (2013) 260:26842690

induces not only hyperplasia (characteristic of CMT1), but also denite proliferation of Schwann cells. A predisposition to develop peripheral and central nervous system tumours (meningiomas, gliomas, and schwannomas) is typical of Neurobromatosis type 2 (NF2), an autosomal dominant disorder caused by mutations in the NF2 gene encoding merlin, a tumour suppressor protein associated with the cytoskeleton [24]. Bilateral vestibular nerve schwannomas occur in 95 % of patients. A peripheral polyneuropathy is frequently seen and can cause substantial disability. It is not clear whether it happens as a separate phenomenon related to merlin dysfunction or as the result of compression of isolated or multiple schwannomas. Different patterns have been reported, i.e. symmetrical sensorimotor polyneuropathy, usually characterised by predominant axonal damage, asymmetric neuropathy with umer focal amyotrophy, and multiple mononeuropathy. Ba and colleagues [24] performed an accurate evaluation of eight NF2 patients (the second largest group reported in the literature) that included an MRI study with high-resolution large-coverage microstructural T2 sampling of upper and lower limbs. They classied nerve abnormalities in non-compressive fascicular microlesions (\ 2 mm), intermediate lesions (25 mm), and compressive macrolesions ([5 mm). The rst two categories were the most frequently found, and their overall burden strongly correlated with neuropathy severity, suggesting that this is the predominant basis of the peripheral nerve damage, i.e. the summation of several intra-nerve micro or intermediate lesions, rather than compression related to macrolesions. These results have an important impact on the decision as to when to undergo surgical removal of macrolesions, and this novel MRI technique offers new investigative possibilities for other diseases characterised by diffuse nerve lesions.

Familial amyloid polyneuropathy Amyloidoses are a heterogeneous group of diseases characterised by localised or systemic tissue deposition of insoluble proteins that form unbranched brillar aggregates of beta-pleated sheet structure termed amyloid. Almost 30 proteins are known to form amyloid brils with several clinical phenotypes that require different diagnostic and therapeutic approaches, depending also on the sites of synthesis of amyloidogenic proteins. In transthyretin-related (ATTR) amyloidosis, the most frequent form of autosomal dominant familial amyloid polyneuropathy (FAP), the deposition of amyloid brils is due to mutations in the transthyretin (TTR) gene coding for the corresponding protein synthesised by liver and, in small amounts, also by choroid plexuses and retina. ATTR amyloidosis usually

manifests with a nerve length-dependent predominantly sensory and autonomic neuropathy, related to the early involvement of small bres; cardiac involvement is frequent and some cases have an exclusive or mainly cardiac phenotype [25]. The most common TTR mutation is p.Val30Met, but more than 100 different mutations are known. Liver transplantation, abolishing the main source of mutated amyloidogenic protein, should be an early consideration in patients with ATTR amyloidosis, especially if they are carrier of the p.Val30Met mutation. Tafamidis meglumine, a TTR stabiliser preventing amyloid deposition, is used for treating early stage symptomatic FAP. Recently, Coelho and colleagues [26] performed an open label extension study and concluded that long-term treatment with tafamidis was well tolerated and reduced the rate of neurologic deterioration over a 30-month period. Pharmacological studies with other TTR stabilisers (Diusinal or a combination of Doxycycline and Taurodesoxycholic acid) are in progress; additionally, gene therapy with small interfering RNAs and antisense oligonucleotides is a new therapeutic perspective for ATTR amyloidosis [27, 28]. Briani and colleagues [29] emphasise the importance of a differential diagnosis between ATTR and other amyloidosis, considering the new promising therapeutic options. They also underline the difculties in diagnosis of these diseases, while describing a patient with restrictive cardiomyopathy, mild IgG-lambda monoclonal gammopathy without neuropathic symptoms, affected by late-onset ATTR amyloidosis associated with the novel p.Glu62Lys TTR mutation. The patient was initially misdiagnosed as being affected by the light-chain (AL) amyloidosis. AL amyloidosis is characterised by systemic involvement and, in about 35 % of cases, peripheral and/or autonomic neuropathy; it is caused by an amyloidogenic accumulation of an immunoglobulin light chain produced by a clonal plasma cell population in the bone marrow. Another important issue is when to start treatment for ATTR amyloidosis, as tissue amyloid deposition starts well before symptoms, and early therapy should allow a better outcome. It is therefore critical to identify early clinical or objective disease manifestations that show that the disease process is ongoing and treatment is warranted. Lefaucheur and coauthors [30] tried to identify early neurophysiological markers of FAP in TTR mutation carriers. In this study, 20 asymptomatic/paucisymptomatic carriers of pathogenic TTR mutations were submitted both to conventional nerve conduction study and small-bre tests including laser evoked potentials, sympathetic skin responses, quantitative sensory testing (QST) with measurement of cold and warm detection thresholds, and heartrate variability assessment. Abnormalities in small-bre tests (more frequently in cold and warm detection

123

J Neurol (2013) 260:26842690

2687

thresholds and laser evoked potentials) were found in 11 of 20 patients (including two asymptomatic carriers), while conventional conduction studies did not show any sign of distal polyneuropathy. Based on their results and because in early TTR-FAP a selective or largely predominant alteration of small bres is documented, the authors suggest that a larger neurophysiological study including combined small-bre tests in TTR mutation carriers can lead to early detection of the rst signs of neuropathy onset. Skin biopsy, by demonstrating decreased intraepidermic nerve bre density, is another potential early marker of disease [31].

to diagnosis in this study was 12.5 years. The authors suggested that lipid prole, with HDL cholesterol and apoA-I measures, should be included in the screening of patients with possible chronic inammatory demyelinating polyneuropathies. Cerebrotendinous Xanthomatosis Cerebrotendinous Xanthomatosis (CTX) is a rare autosomal recessive lipid storage disorder caused by mutations in CYP27A1, the gene encoding the sterol 27-hydroxylase involved in bile acid synthesis, resulting in tissue accumulation of cholestanol, an intermediate compound along the pathway of bile acid synthesis. CTX is characterised by a great variability in disease onset and symptom evolution, even in individuals from the same family. The most common systemic ndings are tendon xanthomas, juvenile cataracts and chronic diarrhoea; neurological features usually occur after the second decade of life and include cognitive impairment, peripheral neuropathy, epilepsy, and cerebello-pyramidal signs. CTX is treatable with chenodeoxycholic acid (CDCA) replacement therapy [34]. Ginanneschi and colleagues [35] examined a large CTX population of 35 patients and analysed the effects of CDCA therapy on polyneuropathy. They showed that the polyneuropathy occurred in 74 % of the cases; it was usually mild, with a sensorimotor or motor axonal polyneuropathy being the most frequent pattern. CDCA treatment improved NCV in all treated patients, by increasing conduction along the still excitable bres; consequently, the authors hypothesised that the treatment promotes myelin synthesis in nerve bres with surviving axons. Fabry disease Fabry disease (FD) is an X-linked lysosomal storage disorder caused by a-galactosidase A gene mutations resulting in accumulation of glycosphingolipids with subsequent organ failure. The most common neuropathic pattern is a small-bre neuropathy that can even be the rst clinical manifestation with onset during childhood or adolescence; further neurological features include cerebrovascular accidents and autonomic dysfunction [36]. Bertelsen and colleagues [37] have recently assessed the presence of smallbre neuropathy in 15 FD patients (ve hemizygous males and ten heterozygous females) with neuropathic pain, by performing clinical examination, QST, corneal confocal microscopy, and skin biopsy. By comparing results of the different methods, the authors concluded that the diagnosis of small-bre neuropathy still relies mainly on clinical symptoms and ndings, and that paraclinical tests can support the diagnosis and are useful for clinical follow-up, as they can document progression of small bre loss.

Neuropathies in metabolic and neurodegenerative diseases Peripheral neuropathy, along with other neurological or systemic symptoms, is a clinical feature of many inherited metabolic or neurodegenerative disorders, which are often misdiagnosed because of their rarity and phenotypic variability. The presence of neuropathy may allow an early diagnosis, which is particularly important because some of these diseases are treatable, e.g., Cerebrotendinous Xanthomatosis and Fabry disease. Furthermore, neuropathy may be a major cause of disability in patients affected by such inherited diseases. Tangier disease Tangier disease (TD) is a rare autosomal recessive disorder caused by mutations in the ATP-Binding Cassette transporter A1 (ABCA1) gene on chromosome 9q31, which encodes the membrane transporter ABCA1 that plays a central role in the transport of cholesterol from peripheral cells to the liver [32]. TD is characterised by intracellular accumulation of cholesterol leading to hepatosplenomegaly, lymphadenopathy, and enlarged yelloworange tonsils, and by low levels of high-density lipoprotein (HDL) cholesterol and apolipoprotein A-I (apoA-I). Approximately 50 % of the patients have a peripheral neuropathy that may present as either a remitting-relapsing mono/polyneuropathy or a syringomyelia-like neuropathy. Zyss and colleagues [33] retrospectively analysed four patients with genetically conrmed TD; three of them had a typical pseudosyringomyelic neuropathy with predominant upper limb weakness together with facial diplegia; the fourth had an atypical asymmetrical onset mimicking LewisSumner syndrome. The authors underscored the clinical variability of disease that, combined with a heterogeneous electrophysiological pattern showing both demyelination and axonal degeneration, may lead to diagnostic errors, especially in the early stages of disease; indeed, average time

123

2688

J Neurol (2013) 260:26842690

POLG1-related neuropathy: MNGIE-like phenotype A predominantly sensory neuropathy, with ataxia and loss of proprioception and vibration sense, is one of the possible presenting features of patients carrying mutations in POLG1, a nuclear gene encoding the mitochondrial DNA polymerase gamma-1. POLG1 mutations can result in different clinical pictures, including progressive external ophthalmoplegia (PEO), sensory ataxic neuropathy with ophthalmoplegia (SANDO), and Alpers syndrome, often with multiple mitochondrial DNA deletions or depletion [38]. Tang and colleagues [39] reported an extremely rare presentation of recessive POLG1 mutations, namely a neurogastrointestinal phenotype, similar to mitochondrial neurogastrointestinal encephalomyopathy (MNGIE). MNGIE is characterised by severe gastrointestinal manifestations, ophthalmoplegia, cachexia, peripheral neuropathy (typically demyelinating) and subclinical leukoencephalopathy, associated with TYMP (thymidine phosphorylase) gene mutations [40]. Tang et al. [39], reviewing clinical features of 92 unrelated patients with recessive POLG1 mutations, noted that three had clinical features consistent with MNGIE, but without leukoencephalopathy. The authors suggest that in MNGIE-like phenotypes, the lack of leukoencephalopathy, in addition to normal plasma thymidine, is suggestive of POLG1 mutations as the responsible molecular defect. Notably, all three patients had a peripheral neuropathy. Allgrove syndrome Vallet and colleagues [41], through a retrospective study of eight adult patients with Allgrove syndrome, described the neurological aspects of this autosomal recessive disease, also known as Triple A syndrome, characterised by achalasia, adrenal insufciency, and alacrima. The authors illustrated the clinical variability of Allgrove syndrome and the diagnostic difculties that arise if neurological symptoms predominate over systemic features, as may occur in late-onset cases. The neurological ndings described in this study are consistent with the previous literature and characterised by pyramidal syndrome, chronic peripheral neuropathy, dysautonomic and bulbar symptoms, and mild cognitive dysfunction. The neuropathy is axonal, predominantly motor, with more severe involvement of ulnar than median nerves. Allgrove syndrome is caused by mutations of the AAAS (AchalasiaAddisonianismAlacrima Syndrome) gene encoding ALADIN, a protein thought to be involved in molecular transport across the nuclear membrane where it is located [42]. In a few individuals with otherwise typical Allgrove syndrome, no AAAS mutation is found, suggesting genetic heterogeneity.

Spastic ataxia of Charlevoix Saguenay Peripheral neuropathy is a typical feature of the autosomal recessive spastic ataxia of Charlevoix Saguenay (ARSACS), a neurodegenerative disorder characterised also by progressive cerebellar ataxia and lower limb spasticity. Although many patients showed the classical ARSACS triad, each of these ndings may be missing and a few patients may present with a pure neuropathy. It is controversial whether the neuropathy is axonal or demyelinating, but all recent series describe a sensory-motor polyneuropathy with reduced nerve conduction velocity (NCV) values [4345]. The underlying pathology appears to be a severe reduction of large myelinated bres, which might account for the reduced NCV, notwithstanding the lack of clear signs of demyelination. ARSACS, rst described in patients from Canadian Quebec, usually starts during early childhood, although adult-onset cases have been described. The disease is due to mutations of the SACS gene encoding sacsin, a protein widely expressed throughout the brain. Recently, it has been shown that SACS mutations alter the mitochondrial network and sacsin seems to be involved in mitochondrial ssion by interacting with dynamin-related protein 1 [46]. Gazulla and coauthors [45] extensively evaluated ve ARSACS patients and proposed a pathogenic mechanism different from neurodegeneration. MRI showed that all ve patients hadbeyond atrophy of the cerebellum, cerebral cortex, and upper cervical corda hypointense pontine linear striation, corresponding to an increased amount of pontocerebellar bres that interrupt the pyramidal tracts. The authors interpreted these results as suggestive of a developmental origin of the disease with pontocerebellar bres compressing the pyramidal tracts at the pons from the embryonic period, thereby causing spasticity from a very early age. The authors conrmed the increased thickness of the retinal nerve bre layer and noticed a straight dorsal spine, elements supportive of a developmental defect. Prodi and coauthors [47] substantiated the MRI nding of hypointense pontine linear striation, but attributed the hypointensity to the cortico-spinal tract being squeezed by the over-represented transverse pontine bres, which are hyperintense on T2-weighted images. They also detected thinning of the corpus callosum, a rim of T2-hyperintensity around the thalami, and microstructural alterations in the supratentorial white matter of forceps, cingulum and superior longitudinal fasciculus in all ARSACS patients analysed, so they suggest that these MRI ndings should be considered diagnostic of ARSACS.
Conicts of interest Dr. Davide Pareyson received research funding from ACMT-Rete and Pzer Italia, and funded travels from Kedrion SpA and Pzer Italia. He is a member of the Inherited

123

J Neurol (2013) 260:26842690 Neuropathy Consortium Rare Disease Clinical Research Consortium funded by the US National Institutes of Health (NINDS/ORD), Muscular Dystrophy Association, and Charcot-Marie-Tooth Association (CMTA). Dr. Giuseppe Piscosquito received funded travels from Pzer Italia.

2689 14. Houlden H, Laura M, Ginsberg L, Jungbluth H, Robb SA, Blake J, Robinson S, King RH, Reilly MM (2009) The phenotype of Charcot-Marie-Tooth disease type 4C due to SH3TC2 mutations and possible predisposition to an inammatory neuropathy. Neuromuscul Disord 19:264269 15. Lewis RA, Sumner AJ, Shy ME (2000) Electrophysiological features of inherited demyelinating neuropathies: a reappraisal in the era of molecular diagnosis. Muscle Nerve 23:14721487 16. Miki Y, Tomiyama M, Haga R, Nishijima H, Suzuki C, Kurihara A, Sugimoto K, Hashiguchi A, Takashima H, Baba M (2013) A family with IVIg-responsive CharcotMarieTooth disease. J Neurol 260:11471151 17. Sugimoto T, Ochi K, Hosomi N, Takahashi T, Ueno H, Nakamura T, Nagano Y, Maruyama H, Kohriyama T, Matsumoto M (2013) Ultrasonographic nerve enlargement of the median and ulnar nerves and the cervical nerve roots in patients with demyelinating Charcot-Marie-Tooth disease: distinction from patients with chronic inammatory demyelinating polyneuropathy. J Neurol. doi:10.1007/s00415-013-7021-0 hr M (2012) Inammatory demye18. Weishaupt JH, Ganser C, Ba linating CNS disorder in a case of X-linked Charcot-Marie-Tooth disease: positive response to natalizumab. J Neurol 259:19671969 19. Scherer SS, Kleopa KA (2012) X-linked Charcot-Marie-Tooth disease. J Peripher Nerv Syst 17:913 20. Kwon JY, Chung KW, Park EK, Park SW, Choi BO (2009) Charcot-Marie-Tooth 1A concurrent with schwannomas of the spinal cord and median nerve. J Korean Med Sci 24:763766 tsch M, Buslei R (2007) Hereditary neuropathy 21. Heckmann JG, Du with liability to pressure palsy combined with schwannomas of the median and medial plantar nerves. Muscle Nerve 35:122124 22. Toyota K, Ogino D, Hayashi M, Taki M, Saito K, Abe A, Hashimoto T, Umetsu K, Tsukaguchi H, Hayasaka K (2013) INF2 mutations in Charcot-Marie-Tooth disease complicated with focal segmental glomerulosclerosis. J Peripher Nerv Syst 18:9798 23. Ohyagi M, Sanjo N, Yokota T, Mizusawa H (2012) Hereditary neuropathy with liability to pressure palsy combined with suspected schwannomas of the peroneal and radial nerves. J Neurol 259:977979 umer T, Schuhmann MU, Tatagiba M, umer P, Mautner VF, Ba 24. Ba Heiland S, Kaestel T, Bendszus M, Pham M (2013) Accumulation of non-compressive fascicular lesions underlies NF2 polyneuropathy. J Neurol 260:3846 -Bordeneuve V, Said G (2011) Familial amyloid polyneu25. Plante ropathy. Lancet Neurol 10:10861097 -Bordeneuve 26. Coelho T, Maia LF, da Silva AM, Cruz MW, Plante V, Suhr OB, Conceic ao I, Schmidt HH, Trigo P, Kelly JW, La` re R, Chan J, Packman J, Grogan DR (2013) Long-term baudinie effects of tafamidis for the treatment of transthyretin familial amyloid polyneuropathy. J Neurol. doi:10.1007/s00415-0137051-7 27. Adams D (2013) Recent advances in the treatment of familial amyloid polyneuropathy. Ther Adv Neurol Disord 6:129139 28. Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, Perez J, Chiesa J, Warrington S, Tranter E, Munisamy M, Falzone R, Harrop J, Cehelsky J, Bettencourt BR, Geissler M, Butler JS, Sehgal A, Meyers RE, Chen Q, Borland T, Hutabarat RM, Clausen VA, Alvarez R, Fitzgerald K, Gamba-Vitalo C, Nochur SV, Vaishnaw AK, Sah DW, Gollob JA, Suhr OB (2013) Safety and efcacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med 369:819829 29. Briani C, Cavallaro T, Ferrari S, Taioli F, Calamelli S, Verga L, Adami F, Fabrizi GM (2012) Sporadic transthyretin amyloidosis with a novel TTR gene mutation misdiagnosed as primary amyloidosis. J Neurol 259:22262228

References
1. Pareyson D, Marchesi C (2009) Diagnosis, natural history, and management of Charcot-Marie-Tooth disease. Lancet Neurol 8:654667 2. Saporta MA, Shy ME (2013) Inherited peripheral neuropathies. Neurol Clin 31:597619 L, Windpassinger C, Bernert G, 3. Fischer C, Trajanoski S, Papic ttl M, Arslan-Kirchner M, Javaher-HagFreilinger M, Schabhu scher WN, Pieber highi P, Plecko B, Senderek J, Rauscher C, Lo TR, Janecke AR, Auer-Grumbach M (2012) SNP array-based whole genome homozygosity mapping as the rst step to a molecular diagnosis in patients with Charcot-Marie-Tooth disease. J Neurol 259:515523 4. Davidson GL, Murphy SM, Polke JM, Laura M, Salih MAM, Muntoni F, Blake J, Brandner S, Davies N, Horvath R, Price S, Donaghy M, Roberts M, Foulds N, Ramdharry G, Soler D, Lunn MP, Manji H, Davis MB, Houlden H, Reilly MM (2012) Frequency of mutations in the genes associated with hereditary sensory and autonomic neuropathy in a UK cohort. J Neurol 259:16731685 5. Rotthier A, Baets J, De Vriendt E, Jacobs A, Auer-Grumbach M, vy N, Bonello-Palot N, Kilic SS, Weis J, Nascimento A, Le Swinkels M, Kruyt MC, Jordanova A, De Jonghe P, Timmerman V (2009) Genes for hereditary sensory and autonomic neuropathies: a genotype-phenotype correlation. Brain 132:26992711 6. Rotthier A, Baets J, Timmerman V, Janssens K (2012) Mechanisms of disease in hereditary sensory and autonomic neuropathies. Nat Rev Neurol 8:7385 7. Coen K, Pareyson D, Auer-Grumbach M, Buyse G, Goemans N, ` M, Scaioli V, Salmhofer W, Claeys KG, Verpoorten N, Laura Pieber TR, Nelis E, De Jonghe P, Timmerman V (2006) Novel mutations in the HSN2 gene causing hereditary sensory and autonomic neuropathy type II. Neurology 66:748751 lez-Huerta LM, Valde s-Miranda JM, 8. Pacheco-Cuellar G, Gonza ez-Gonza lez H, Zenteno-Bacheron S, Cazarin-Barrientos J, Pela Cuevas-Covarrubias SA (2011) Hereditary sensory and autonomic neuropathy II due to novel mutation in the HSN2 gene in Mexican families. J Neurol 258:18901892 9. Shekarabi M, Dion PA, Rouleau GA (2012) Letter to the editors: comment on hereditary sensory and autonomic neuropathy II due to novel mutation in the HSN2 gene in Mexican families. J Neurol 259:565566 10. Marttila M, Rautenstrauss B, Huehne K, Laitinen V, Majamaa K, rppa M (2012) A novel mutation of myelin protein zero Ka associated with late-onset predominantly axonal Charcot-MarieTooth disease. J Neurol 259:15851589 11. Shy ME, Jani A, Krajewski K, Grandis M, Lewis RA, Li J, Shy RR, Balsamo J, Lilien J, Garbern JY, Kamholz J (2004) Phenotypic clustering in MPZ mutations. Brain 127:371384 ` M, Milani M, Morbin M, Moggio M, Ripolone M, Jann S, 12. Laura Scaioli V, Taroni F, Pareyson D (2007) Rapid progression of late onset axonal Charcot-Marie-Tooth disease associated with a novel MPZ mutation in the extracellular domain. J Neurol Neurosurg Psychiatry 78:12631266 13. Ginsberg L, Malik O, Kenton AR, Sharp D, Muddle JR, Davis MB, Winer JB, Orrell RW, King RH (2004) Coexistent hereditary and inammatory neuropathy. Brain 127:193202

123

2690 30. Lefaucheur JP, Ng Wing Tin S, Kerschen P, Damy T, Plante Bordeneuve V (2013) Neurophysiological markers of small bre neuropathy in TTR-FAP mutation carriers. J Neurol 260: 14971503 31. Joint Task Force of the EFNS and the PNS (2010) European Federation of Neurological Societies/Peripheral Nerve Society Guideline on the use of skin biopsy in the diagnosis of small ber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. J Peripher Nerv Syst 15:7992 32. Rust S, Rosier M, Funke H, Real J, Amoura Z, Piette JC, Deleuze ` e P, Assmann G (1999) JF, Brewer HB, Duverger N, Dene Tangier disease is caused by mutations in the gene encoding ATP-binding cassette transporter 1. Nat Genet 22:352355 hin A, Couvert P, Bouhour F, Sassolas A, Kolev I, 33. Zyss J, Be A, Stojkovic T (2012) Clinical Denys V, Vial C, Lacour A, Carrie and electrophysiological characteristics of neuropathy associated with Tangier disease. J Neurol 259:12221226 rkhem I (2013) Cerebrotendinous xanthomatosis. Curr Opin 34. Bjo Lipidol 24:283287 35. Ginanneschi F, Mignarri A, Mondelli M, Gallus GN, Del Puppo M, Giorgi S, Federico A, Rossi A, Dotti MT (2013) Polyneuropathy in cerebrotendinous xanthomatosis and response to treatment with chenodeoxycholic acid. J Neurol 260:268274 36. Bersano A, Lanfranconi S, Valcarenghi C, Bresolin N, Micieli G, Baron P (2012) Neurological features of Fabry disease: clinical, pathophysiological aspects and therapy. Acta Neurol Scand 126:7797 37. Bertelsen AK, Tndel C, Krohn J, Bull N, Aarseth J, Houge G, Mellgren SI, Vedeler CA (2013) Small bre neuropathy in Fabry disease. J Neurol 260:917919 38. Milone M, Massie R (2010) Polymerase gamma 1 mutations: clinical correlations. Neurologist 16:8491 39. Tang S, Dimberg EL, Milone M, Wong LJ (2012) Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE)-like phenotype: an expanded clinical spectrum of POLG1 mutations. J Neurol 259:862868

J Neurol (2013) 260:26842690 40. Pareyson D, Piscosquito G, Moroni I, Salsano E, Zeviani M (2013) Peripheral neuropathy in mitochondrial disorders. Lancet Neurol. doi:10.1016/S1474-4422(13)70158-3 41. Vallet AE, Verschueren A, Petiot P, Vandenberghe N, Nicolino M, Roman S, Pouget J, Vial C (2012) Neurological features in adult Triple-A (Allgrove) syndrome. J Neurol 259:3946 42. Houlden H, Smith S, De Carvalho M, Blake J, Mathias C, Wood NW, Reilly MM (2002) Clinical and genetic characterization of families with triple A (Allgrove) syndrome. Brain 125:26812690 43. Synofzik M, Soehn AS, Gburek-Augustat J, Schicks J, Karle KN, ning M, Biskup S, Rudnik-Scho neborn le R, Haack TB, Scho Schu S, Senderek J, Hoffmann KT, MacLeod P, Schwarz J, Bender B, ls L (2013) Autosomal reces ger S, Kreuz F, Bauer P, Scho Kru sive spastic ataxia of Charlevoix Saguenay (ARSACS): expanding the genetic, clinical and imaging spectrum. Orphanet J Rare Dis 8:41 44. Stevens JC, Murphy SM, Davagnanam I, Phadke R, Anderson G, Nethisinghe S, Bremner F, Giunti P, Reilly MM (2013) The ARSACS phenotype can include supranuclear gaze palsy and skin lipofuscin deposits. J Neurol Neurosurg Psychiatry 84:114116 n MA, Pablo LE, Tessa A, 45. Gazulla J, Benavente I, Vela AC, Mar M, Barrena MR, Santorelli FM, Nesti C, Modrego P, Tintore Berciano J (2012) New ndings in the ataxia of CharlevoixSaguenay. J Neurol 259:869878 ` re R, Partt DA, Deane EC, Gaudet R, Nossova 46. Girard M, Larivie N, Blondeau F, Prenosil G, Vermeulen EG, Duchen MR, Richter A, Shoubridge EA, Gehring K, McKinney RA, Brais B, Chapple JP, McPherson PS (2012) Mitochondrial dysfunction and Purkinje cell loss in autosomal recessive spastic ataxia of CharlevoixSaguenay (ARSACS). Proc Natl Acad Sci USA 109:16611666 47. Prodi E, Grisoli M, Panzeri M, Minati L, Fattori F, Erbetta A, Uziel G, DArrigo S, Tessa A, Ciano C, Santorelli FM, Savoiardo M, Mariotti C (2012) Supratentorial and pontine MRI abnormalities characterize recessive spastic ataxia of CharlevoixSaguenay. A comprehensive study of an Italian series. Eur J Neurol 20:138146

123

You might also like