Abstract
Multiple system atrophy (MSA) is a rare neurodegenerative disorder without any effective treatment in slowing or stopping disease progression. It is characterized by poor levodopa responsive Parkinsonism, cerebellar ataxia, pyramidal signs and autonomic failure in any combination. Current therapeutic strategies are primarily based on dopamine replacement and improvement of autonomic failure. However, symptomatic management remains disappointing and no curative treatment is yet available. Recent experimental evidence has confirmed the key role of alpha-synuclein aggregation in the pathogenesis of MSA. Referring to this hypothesis, transgenic and toxic animal models have been developed to assess candidate drugs for MSA. The standardization of diagnosis criteria and assessment procedures will allow large multicentre clinical trials to be conducted. In this article we review the available symptomatic treatment, recent results of studies investigating potential neuroprotective drugs, and future approaches for the management in MSA.
Keywords: multiple system atrophy, neurodegenerative diseases, neuroprotective agents, Parkinsonism, therapeutic
Introduction
Multiple system atrophy (MSA) is a progressive neurodegenerative disorder leading to more severe disability and impairment in quality of life than in Parkinson’s disease (PD) [Schrag et al. 2006; Tison et al. 2002]. MSA is characterized by a variable combination of poor levodopa responsive Parkinsonism, cerebellar ataxia and autonomic failure [Gilman et al. 2008]. MSA-Parkinsonism type (MSA-P) predominates in the Western Hemisphere and MSA-cerebellar type (MSA-C) is the major phenotype in the Eastern Hemisphere [Watanabe et al. 2002]. There is no recognized efficient treatment for cerebellar ataxia whereas levodopa may transiently improve Parkinsonism in one third of the patients [Constantinescu et al. 2007; Wenning et al. 1997]. Since autonomic failure strongly correlates with poor quality of life [Schrag et al. 2006], therapeutic management focuses on symptomatic therapy of orthostatic hypotension (OH), constipation, genitourinary and breathing disorders. Mean survival is about 9–10 years after symptom onset [Schrag et al. 2008] with nocturnal sudden death being a major cause of death [Shimohata et al. 2008]. Hitherto, no treatment has been shown to improve survival in MSA, creating a strong need for new therapeutic approaches.
Abnormal aggregation of alpha-synuclein in oligodendrocytes is the pathological hallmark of MSA and may trigger the progressive cell loss in widespread brain areas [Jellinger and Lantos, 2010; Wenning et al. 2008, 1997]. Advances in therapeutics have been achieved during the past 10 years through the development of transgenic animal models and experimental strategies against alpha-synuclein accumulation and for neuroprotection [Stefanova et al. 2009]. This progress may provide a testbed for future candidate drugs discovery and multicentric clinical trials.
Here we review potential neuroprotective drugs, current symptomatic treatment and future approaches in the management of MSA.
Current therapies
A list of the currently symptomatic treatment for MSA is given in Table 1.
Table 1.
Feature | Current first-line treatment | Alternative treatment |
---|---|---|
Parkinsonism | Levodopa up to 1 g/day if tolerated, in association with domperidone and physiotherapy | Dopamine agonist, amantadine, paroxetine [Friess et al. 2006] |
Cerebellar ataxia | Physiotherapy | Clonazepam, propanolol, baclofen, amantadine, gabapentin, buspirone |
Orthostatic hypotension | Nonpharmacological measures (custom-fitted elastic stockings, raising the head of the bed when sleeping, water drinking, small meals). Midodrine from 2.5 to 30 mg per day [Wright et al. 1998; Low et al. 1997; Jankovic et al. 1993]a | Fludrocortisone, pyridostigmine,b droxidopa [Mathias, 2008],c [Kaufmann et al. 2003], antidiuretic hormone desmopressin at bedtime, ephedrine, octreotide |
Neurogenic lower urinary tract dysfunction | If postvoid residual >100 ml, clean intermittent catheterization | Botulinum toxin A in the detrusor muscle or the urethral sphincter, surgery options, permanent catheterization |
If postvoid residual <100 ml, anticholinergic agents for detrusor hyperactivity, α-adrenergic antagonists for urethra hypertony | ||
Constipation | High fluid and fibre intake classical laxative therapy, polycarbophil, mosapride citrate, macrogol 3350 | |
Erectile dysfunction | Intracavernosal injection of papaverine or prostaglandin E1, sildenafil [Hussain et al. 2001] | Subcutaneous apomorphine injections |
Breathing disorders | Continuous positive air pressure | Tracheostomy (for life-threatening and/or daytime stridor, abnormal vocal cord mobility on laryngoscopy), laryngeal surgery or botulinum toxin injections, adaptive servoventilation |
Dystonia | Botulinum toxin injection, levodopa therapy | Anticholinergics, amantadine, dopamine agonists, muscle relaxants, tetrabenazine |
Camptocormia | Physiotherapy with specific orthosis, wearing a backpack | Botulinum toxin injections, protirelin tartrate |
Rapid eye movement sleep behavior disorder | Clonazepam | Sodium oxybate, temazepam, zopiclone, gabapentin, pramipexole, donepezil, melatonine |
Depression | Psychotherapy | Electroconvulsive therapy |
Selective serotonergic reuptake inhibitors, levodopa therapy | Repetitive transcranial magnetic stimulation | |
Cognitive impairment | Speech therapy | |
Drooling | Anticholinergic drugs | Injection of botulinum toxin into the salivary glands |
Among all of the subjects included in these trials, there was a minority of MSA patients: 40/171 subjects [Low et al. 1997], 7/25 [Wright et al. 1998], and 18/97 [Jankovic et al. 1993]. In available subgroup analysis, beneficial effect on orthostatic hypotension (OH) was reported [Low et al. 1997; Jankovic et al. 1993].
Positive effect on OH regarding subgroup analysis in 17 MSA patients of the 58 subjects included, but there was no placebo group control [Singer et al. 2006].
Preliminary results.
Parkinsonism
A poor levodopa response is one of the consensus criteria for the diagnosis of MSA [Gilman et al. 2008] and helps to differentiate MSA from PD [Wenning et al. 2000]. Although there are no controlled clinical trials on the efficacy of levodopa in MSA, about one third of patients may experience a benefit, more often in the MSA-P subtype than in MSA-C [Constantinescu et al. 2007]. In series with pathological confirmation, the positive levodopa responsiveness is reported in 28–65% of the patients [Colosimo et al. 1995; Wenning et al. 1997, 1995; Hughes et al. 1992; Fearnley and Lees, 1990; Rajput et al. 1990]. However, this effect persists for several years (as in PD) in only 13% of all patients [Wenning et al. 1994]. Although levodopa induces less delirium and hallucinations in MSA than in PD [Wenning et al. 2000], it can lead to adverse effects such as worsening of OH or pathological hypersexuality [Klos et al. 2005; Wenning et al. 1994]. Moreover, early orofacial levodopa induced dyskinesias may occur in half of MSA patients often in the absence of any motor benefit [Boesch et al. 2002; Wenning et al. 1994]. According to consensus criteria, levodopa unresponsiveness should only be accepted after a treatment with at least 1 g of levodopa per day for at least 3 months [Gilman et al. 2008]. However, some patients report a subjective benefit that may not be apparent in a standardized motor examination, while others convey subsequent motor degradation after levodopa discontinuation [Wenning et al. 1994]. To date, there is no evidence suggesting that such high doses of levodopa accelerate the neurodegenerative process in transgenic animal models of MSA [Stefanova et al. 2007]. As in PD, domperidone can be added to prevent dopaminergic side effects such as nausea and vomiting.
There are no controlled studies that have tested the efficacy of dopamine agonists in MSA so far. In a retrospective case record analysis, only 10% of the patients experienced a benefit with dopamine agonists [Wenning et al. 1994]. Thus, dopamine agonists cannot be recommended as a first-line treatment given their high rate of side effects compared with levodopa, particularly worsening of OH. Amantadine may be an alternative symptomatic treatment. While a double-blind, placebo-controlled crossover trial failed to demonstrate any significant motor benefit in eight MSA patients [Wenning, 2005], a retrospective study disclosed good or excellent responsiveness in 15% [Wenning et al. 1994].
Although serotonergic involvement in MSA remains unclear, postmortem studies disclosed clear evidence for serotonergic depletion in brainstem nuclei [Tada et al. 2009; Benarroch, 2007]. The selective serotonin reuptake inhibitor paroxetine has been assessed for 2 weeks in a small trial including 19 MSA patients [Friess et al. 2006]. In this double-blind, placebo-controlled, randomized study, motor improvement was slight but significant in comparison with placebo. Further investigations are needed before any conclusion can be drawn on the efficacy of selective serotonin reuptake inhibitors in MSA. A double-blind, multicentre trial with fluoxetine is currently underway in France.
Cerebellar ataxia
Physiotherapy remains the best therapeutic option for cerebellar ataxia in MSA [Jain et al. 2004]. When intentional cerebellar tremor predominates, off-label use of low doses of clonazepam may sometimes help in our hands. Off-label use of propanolol, baclofen or amantadine have shown modest and transient efficacy in a retrospective data analysis [Wenning et al. 2003]. Buspirone (off-label) improved upper limb ataxia in an open-label trial including nine MSA-C patients [Heo et al. 2008]. Gabapentin (off-label) may have symptomatic benefits on ataxia, oscillopsia and dysarthria according to a limited report in two MSA-C patients [Gazulla and Benavente, 2005]. Finally, protirelin tartrate (off-label) has been used for ataxic diseases in Japan, but without any apparent effect in MSA [Takei et al. 2009].
Autonomic failure
Orthostatic hypotension
OH is one of the major criteria for MSA diagnosis [Gilman et al. 2008]. Spreading of the neurodegenerative process to the intermediolateral cell column [Wenning et al. 1997] underlies earlier and more severe autonomic failure in MSA compared with PD [Lipp et al. 2009]. First-line treatment should always include nonpharmacological advice: use of custom-fitted elastic stockings, raising the head of the bed by 20–30° when sleeping, performing isotonic exercises, as well as increasing intake of fluid, salt and frequency of small meals [Freeman, 2008]. Drinking more than 350 ml of water a day reduces both OH and orthostatic symptoms but long-term effects and acceptance are unknown [Deguchi et al. 2007; Young and Mathias, 2004].
A broad range of drugs has been assessed in OH, such as fludrocortisone, midodrine, ephedrine or octreotide [Wenning et al. 2003]. Only midodrine has been investigated with a strong, double-blind, placebo-controlled procedure in three studies including few patients with MSA and labelled in the indication of neurogenic OH [Wright et al. 1998; Low et al. 1997; Jankovic et al. 1993]. This α1-adrenoceptor agonist alleviated moderate to severe OH with oral doses that ranged from 2.5 to 30 mg daily. Blood pressure should be regularly monitored in patients receiving midodrine to detect supine hypertension which may be prevented by taking the last pill more than 4 hours before bedtime [McClellan et al. 1998]. By enhancing sympathetic activity, pyridostigmine modestly but significantly improved OH without supine hypertension in a double-blind, randomized, crossover study [Singer et al. 2006]. Although almost exclusively used in Japan, droxidopa seems to be effective and well tolerated to treat OH in patients with MSA [Kaufmann, 2008; Mathias, 2008; Kaufmann et al. 2003; Mathias et al. 2001; Freeman et al. 1999].
Neurogenic lower urinary tract dysfunction
Urinary urgency, frequency or incomplete bladder emptying, and urinary incontinence remain key features of the consensual diagnosis criteria [Gilman et al. 2008]. Urinary disorders severely affect the quality of life in MSA patients and may lead to lower urinary tract and kidney infection as major causes of morbidity in MSA [Ito et al. 2006]. Clean intermittent self-catheterization (CISC) is recommended as the first-line treatment when the postvoid residual is above 100 ml [Fowler and O’Malley, 2003]. Thus, the residual volume should be regularly monitored, for example with a portable ultrasound device. The critical threshold of 100 ml is reached in a mean of 2 years after disease diagnosis [Ito et al. 2006]. If the postvoid residual is below 100 ml, drugs acting on the detrusor may be used [Papatsoris et al. 2008]. Bladder-oriented anticholinergic agents are often prescribed for detrusor hyperactivity symptoms (e.g. urinary urgency, frequency and urge incontinence) but with a risk of worsening urinary retention. Urethra-orientated α-adrenergic antagonists may reduce the residual urine volume but exacerbation of OH is likely to occur [Sakakibara et al. 2000]. If urge incontinence still persists, third-line treatments may be considered. Synthetic antidiuretic hormone desmopressin (off-label) decreases urinary volumes. Taken just before bedtime, desmopressin reduces nocturia and improves morning hypotension. Regular monitoring of drug safety is necessary since it may induce water intoxication [Sakakibara et al. 2003]. Injection of botulinum toxin A in the detrusor muscle may be an alternative treatment for detrusor overactivity given the positive results in two MSA patients [Giannantoni et al. 2009], as well as urethral sphincter injections for urethral hypertonia when alpha-blockers are not tolerated [Apostolidis et al. 2009]. Botulinum toxin injections (off-label) seem to be safe and effective but further trials with larger samples are needed in MSA. To reduce the risk of infection due to permanent catheterization, surgery such as sphincterotomy or sphincteric wall stenting can be considered as a last option in MSA [Ito et al. 2006; Fowler and O’Malley, 2003].
Constipation
In addition to exercise, high fluid and fibre intake, laxative therapy is often necessary to relieve constipation in MSA. As in PD, classical laxative treatment may be used. Two small open trials have demonstrated the efficacy of polycarbophil [Sakakibara et al. 2007] and mosapride citrate [Liu et al. 2005] to reduce the time of transit in MSA patients. Moreover, macrogol 3350 has been reported to be subjectively efficient in two MSA patients [Eichhorn and Oertel, 2001].
Erectile dysfunction
Intracavernosal injection of papaverine or prostaglandin E1 may be used for erectile dysfunction in MSA [Papatsoris et al. 2008]. Subcutaneous apomorphine administration may also induce erections in PD [O’Sullivan, 2002; O’Sullivan and Hughes, 1998]. Although its use has not been assessed in this indication in MSA patients, apomorphine is easier to handle than papaverine. The use of sildenafil in MSA often faces the problem of its cardiovascular side effects, mainly the risk of a severe blood pressure drop. Efficiency and safety of the latter was assessed in a randomized, double-blind, placebo-controlled, crossover study [Hussain et al. 2001]. Sildenafil enhanced erectile function in the six MSA patients studied, but half of them experienced a severe drop in blood pressure 1 hour after drug ingestion. The pre-existing severity of OH may be predictive for a further decrease in blood pressure and the appearance of clinical signs of orthostatism.
Breathing disorders
MSA may manifest with obstructive and central sleep apnoea, dysrhythmia, dyspnoea, hypoxaemia, inspiratory sighs and laryngeal stridor [Meissner et al. 2010; Gilman et al. 2008; Shimohata et al. 2007; Geser et al. 2005; Vetrugno et al. 2004; Chokroverty et al. 1978]. These breathing disorders reflect the extension of the neurodegenerative process to the pontomedullary respiratory nuclei [Benarroch, 2007]. Since respiratory disorders may manifest in the early stages of MSA [Glass et al. 2006], and may explain sudden death [Tada et al. 2009], they should be considered as a primordial aspect of management in MSA. Stridor is usually the consequence of vocal cord dystonia [Vetrugno et al. 2007; Merlo et al. 2002] but cases with vocal cord abduction muscle denervation have been documented [Hayashi et al. 1997; Bannister et al. 1981]. Its occurrence is known to be associated with poor outcome [Yamaguchi et al. 2003; Silber and Levine, 2000]. Invasive procedures should be considered after the failure of noninvasive ventilation. Tracheostomy may be discussed with the patient for life-threatening and/or daytime stridor, or when abnormal vocal cord mobility is diagnosed on laryngoscopic examination [Silber, 2008; Silber and Levine, 2000]. However, the latter do not seem to prevent sudden death [Shimohata et al. 2008; Jin et al. 2007]. The use of laryngeal surgery or botulinum toxin therapy is sometimes performed with contrasting effects in limited reports [Iranzo, 2005; Merlo et al. 2002].
Concerning obstructive sleep apnoea, treatment with continuous positive air pressure may reduce the apnoea/hypopnoea index and desaturation [Iranzo et al. 2004, 2000], but we found the acceptability to be low when disease progresses [Ghorayeb et al. 2005b]. Moreover, the mean survival does not seem to be modified according to retrospective and/or uncontrolled data [Shimohata et al. 2008; Ghorayeb et al. 2005b]. Finally, recent clinical evidence suggests that adaptive servoventilation may reduce the apnoea index in MSA patients with central sleep apnoea [Suzuki et al. 2010].
Movement disorders
Orofacial dystonia, antecollis and camptocormia are features supporting the consensual diagnosis of MSA and constitute ‘red flags’ for the differential diagnosis with PD [Gilman et al. 2008; Kollensperger et al. 2008].
Dystonia
In a prospective trial including 24 levodopa-naive patients with probable MSA, dystonia occurred in 46% [Boesch et al. 2002]. Botulinum toxin injection is the most commonly used treatment for focal dystonia [Papapetropoulos et al. 2008]. Although no controlled studies with any other drug are yet available, symptomatic relief with anticholinergics, amantadine, dopamine agonists, muscle relaxants or tetrabenazine has been reported in some MSA patients when used off-label [Papapetropoulos et al. 2008; Azher and Jankovic, 2005].
Camptocormia
Camptocormia (CC) is an abnormal forward flexion of the trunk that manifests when standing or walking and relieves in the supine position [Azher and Jankovic, 2005]. Whether CC is a dystonic posture, a consequence of axial rigidity or a focal myopathy is still debated [Margraf et al. 2010; Diederich et al. 2006; Slawek et al. 2006; Azher and Jankovic, 2005]. The levodopa responsiveness is generally poor [Bloch et al. 2006; Diederich et al. 2006; Azher and Jankovic, 2005], but some patients reported fair relief [Song et al. 2008; Slawek et al. 2006]. In a single case report, protirelin tartrate (off-label use) improved CC dramatically, presumably by enhancing motoneuronal excitability of the paraspinal muscles [Takei et al. 2009]. As in focal dystonia, botulinum toxin injections may be effective [Azher and Jankovic, 2005]. Despite the lack of placebo-controlled trials, injections may be more appropriate in rectus abdominus muscles than in lumbar paraspinal muscles because of the risk of transient aggravation of CC. Finally physiotherapy in combination with specific orthosis may improve CC and quality of life [de Seze et al. 2008]. Recent clinical evidence suggests that wearing a backpack may also alleviate CC in PD [Gerton et al. 2010]. Although the efficacy has not been specifically tested in MSA, such noninvasive options should be considered.
Rapid eye movement sleep behaviour disorder
Rapid eye movement sleep behaviour disorder (RBD) may be the presenting symptom of MSA [Tison et al. 1995] and is observed in 69–100% of systematic polysomnography recordings in MSA patients [Vetrugno et al. 2004; Plazzi et al. 1997; Manni et al. 1993]. Although no therapeutic strategy has been validated, the off-label use of clonazepam is consensually and by experience considered as the first-line treatment [Ghorayeb et al. 2005a]. Clonazepam may aggravate existing obstructive sleep apnoea, but alternative treatments are sparse. Sodium oxybate, temazepam, zopiclone, gabapentin and pramipexole were effective when used off-label in limited and uncontrolled single case reports [Anderson and Shneerson, 2009]. Donepezil, a centrally acting reversible acetylcholinesterase inhibitor, is expected to alleviate RBD (off-label use) [Ringman and Simmons, 2000], but its reported clinical efficacy is variable [Boeve et al. 2003]. Melatonin may restore RBD-related desynchronization of the circadian rhythms without major side effects [Boeve et al. 2003], but these data await confirmation.
Nonspecific therapies
Depression
Depression is more prevalent in MSA than in PD [Tison et al. 2006] and correlates with poor quality of life [Schrag et al. 2006]. Selective serotonergic reuptake inhibitors are the most prescribed antidepressive treatment, with less expected risk of inducing OH than tricyclic drugs. Electroconvulsive therapy may be considered in MSA patients with melancholia [Shioda et al. 2006]. Repetitive transcranial magnetic stimulation may also have antidepressant efficacy in PD but the effects in MSA are unknown [Fregni et al. 2004]. Psychological support is necessary, as well as nursing care and family education [Hardy, 2008]. Finally, levodopa may slightly improve mood disorders in MSA [Fetoni et al. 1999].
Cognitive impairment
MSA patients may have impaired visuospatial and constructional function, verbal fluency and executive function compared with control subjects [Kitayama et al. 2009; Kawai et al. 2008]. No specific cognition enhancement treatment has been assessed in MSA.
Medical rehabilitation
For cerebellar ataxia and Parkinsonism, medical rehabilitation seems to improve balance and motor impairment [Landers et al. 2009; Wedge, 2008; Jain et al. 2004] as does the practice of tai chi [Venglar, 2005]. Speech therapy may be necessary to improve communication because of severe dysarthria.
Drooling
Excessive drooling may be a problem in later disease stages. Anticholinergic drugs may be efficient but adverse effects are frequent, including dry mouth, cognitive impairment, constipation, blurred vision or urinary retention. Injection of botulinum toxin type A into the salivary glands is the treatment of choice in excessive drooling as central side effects can be avoided [Mancini et al. 2003]. However, it may aggravate swallowing problems which may require transient nasogastric tube feeding.
Neuroprotective strategies
Although recent advances in basic science have given some clues for neuroprotective strategies in MSA patients, all clinical trials failed to show any disease-modifying properties (Table 2). Experimental evidence in a rodent model of MSA suggested that the antiglutamate drug riluzole may delay neuronal loss [Diguet et al. 2005]. No positive effect was noted in two prospective trials performed in MSA, using validated clinical scales and survival time as outcomes [Bensimon et al. 2009; Seppi et al. 2006].
Table 2.
Putative neuroprotective drugs that have been assessed in MSA | Putative neuroprotective drugs under clinical assessment |
---|---|
Riluzole [Bensimon et al. 2009; Seppi et al. 2006] | Lithium |
Minocycline [Dodel et al. 2010] | Rasagiline |
Growth hormone [Holmberg et al. 2007] | Intravenous immunoglobulins |
Oestrogen [Heo et al. 2008] |
To test the hypothesis that minocycline inhibits microglial activation, which is supposed to contribute to the progressive cell death in MSA, a 48-week prospective study was performed in 63 MSA-P patients [Dodel et al. 2010]. Although a subgroup analysis of eight patients revealed a nonsignificant decrease in [11C](R)-PK11195-PET binding, a tracer of glial cells including microglia, astrocytes and infiltrating macrophages, there was no change in clinical measures of motor impairment or health-related quality of life.
Since growth hormone (GH) may also act as a neuronal ‘survival factor’ in MSA, a randomized, double-blind, placebo-controlled trial has been designed with 22 MSA patients receiving recombinant human growth hormone (r-hGH) injection during 1 year and 21 receiving placebo [Holmberg et al. 2007]. Although this study disclosed no significant effect, there was a trend to a smaller increase in Unified Parkinson’s Disease Rating Scale (UPDRS) and Unified Multiple System Atrophy Rating Scale (UMSARS) scores over time as well as in OH and cardiovascular variability. Since the use of r-hGH appears to be safe, further trials with higher doses and more patients are needed.
Finally, in MSA-C patients, the hypothesis that oestrogen could have neuroprotective properties has not been confirmed in a recent trial [Heo et al. 2008].
Deep brain stimulation
Although bilateral subthalamic stimulation may have beneficial effects in a few MSA patients [Visser-Vandewalle et al. 2003], a recent review of the literature highlights the poor efficacy of deep brain simulation (DBS) [Shih and Tarsy, 2007]. Moreover, more than a quarter of patients died within 7 months of surgery. Owing to the limited number of reports, the poor outcome and the possibility of a harmful effect, DBS is currently not recommended in MSA [Wenning and Stefanova, 2009; Lambrecq et al. 2008; Shih and Tarsy, 2007; Santens et al. 2006; Talmant et al. 2006; Tarsy et al. 2003].
Future therapies
Neuroprotective strategies
Although the exact mechanisms of the neurodegenerative process in MSA remain unclear, the aggregation of alpha-synuclein in oligodendrocytes has been identified as a critical step in the pathogenesis [Jellinger and Lantos, 2010; Ubhi et al. 2010; Stefanova et al. 2009; Wenning et al. 2008]. Based on the key role of alpha-synuclein aggregation in MSA, transgenic animal models and genetic strategies have been developed. Transgenic animal models allow the expression of alpha-synuclein in oligodendrocytes under control of specific promoters [Shults et al. 2005; Yazawa et al. 2005; Kahle et al. 2002]. The growing number of MSA animal models [Fernagut et al. 2005; Stefanova et al. 2005] opens up the possibility to create a basis for drug screening in human trials. The efficacy of neuroprotective drugs is assessed in rodent models before translation to clinical trials. Furthermore, transgenic models may be used to understand the alpha-synuclein aggregation process and allow screening for candidate drugs before further assessment in clinical trials [Waxman and Giasson, 2010; Ono et al. 2007].
Lithium is a first-line treatment for bipolar mood disorders [Beaulieu and Caron, 2008]. The set of evidence has grown to suggest that lithium may also have also some neuroprotective properties [Ferrucci et al. 2010; Beaulieu and Caron, 2008; Feng et al. 2008; Fornai et al. 2008]. In human pathology, lithium has been tried in amyotrophic lateral sclerosis [Bedlack et al. 2008; Fornai et al. 2008; Vanacore and Galeotti, 2008]. The safety, tolerability and efficacy of lithium are being assessed in MSA in a prospective phase 2 trial (ClinicalTrials.gov Identifier: NCT00997672).
The results of a recent trial suggest that rasagiline may have ‘disease-modifying’ properties of still uncertain clinical significance in PD patients [Olanow et al. 2009b]. Positive effects of rasagiline on clinical motor scores and neuronal loss have also been suggested in a transgenic model of MSA [Stefanova et al. 2008]. Rasagiline is currently being assessed in MSA-P patients in a randomized, placebo-controlled, multicentre study (ClinicalTrials.gov Identifier: NCT00977665).
Nonsteroidal anti-inflammatory drugs (NSAIDs) reduce alpha-synuclein fibril formation in vitro [Hirohata et al. 2008] and may lower the risk of developing PD [Samii et al. 2009; Chen et al. 2005]. To date, the potential therapeutic benefit of NSAIDs has not been assessed in symptomatic alpha-synucleinopathies such as MSA [Hirsch and Hunot, 2009]. However, the harmful adverse effects of long-term use may limit their evaluation until the development of safer drugs. No clinical trials have yet been conducted or planned in MSA.
An open trial testing intravenous immunoglobulins for 6 months is currently in phase 2 (ClinicalTrials.gov Identifier: NCT00750867). However, a former clinical report does not support any therapeutic effect in MSA [Nanri et al. 2009].
Since rifampicin avoids alpha-synuclein fibril formation in vitro [Ono and Yamada, 2006] and reduces neuronal loss in a rodent model of MSA [Ubhi et al. 2008], this drug may be a candidate neuroprotective agent in MSA. However, to the best of the authors’ knowledge, no human trial has been yet planned.
Neurorestorative approach
Although experimental and open trials may support transplantation of foetal dopaminergic cells into the striatum in PD [Hauser et al. 1999; Kordower et al. 1998; Freeman et al. 1995; Widner et al. 1992; Lindvall et al. 1994, 1990], two double-blind, sham-controlled trials assessing transplantation of foetal nigral cells in PD failed to demonstrate any significant effect of the graft considering the primary outcome [Olanow et al. 2003; Freed et al. 2001]. Moreover, off-medication dyskinesia may occur in half of the transplanted patients [Olanow et al. 2009a], and the set of evidence has grown to suggest that Lewy body pathology may affect graft cells [Apostolidis et al. 2009; Kordower et al. 2008a, 2008b]. Despite these shortcomings, recent evidence in animal models of MSA-P may provide the rationale for human trials in MSA [Kollensperger et al. 2009; Puschban et al. 2005]. The use of autologous stem cells has recently been assessed in an open trial with MSA-C patients during 1 year [Lee et al. 2008]. Parenteral injection of bone marrow mesenchymal stem cells seem to slow UMSARS progression and increase cerebellar and frontal glucose metabolism on positron emission tomography (PET) scans compared with nontreated MSA patients. These preliminary results are still to be taken with caution. The underlying mechanisms of action, in particular how graft cells pass the blood–brain barrier from the arterial or venous peripheral circulation, remain unclear and need more preclinical explanation [Whone and Scolding, 2009; Quinn et al. 2008]. A double-blind, placebo-controlled, randomized clinical trial with initial arterial and subsequent venous administration of autologous mesenchymal stem cells has been launched in MSA patients in May 2009 (ClinicalTrials.gov Identifier: NCT00911365).
At the same time, a stem cell boosting strategy with injection of granulocyte colony stimulating factor (GCSF) has been launched in an open uncontrolled trial in four MSA patients [Pezzoli et al. 2010]. No major side effect was noted except for bone pain. However, given the small sample size, it is currently impossible to draw any conclusions about the efficacy of this approach.
Unexplained sudden death
For Schrag and collaborators, therapeutic management should concentrate on autonomic dysfunction, motor impairment and depression, because these features are associated with poor quality of life in MSA [Schrag et al. 2006]. In addition to symptomatic management and neuroprotective strategies, sudden death does not yet represent a major target for therapeutic intervention and there is no trial designed for such a strategy. Occurrence of sudden death is a common cause of mortality in MSA and may happen in the early stages while disability remains acceptable [Shimohata et al. 2008]. The origin of sudden death in MSA remains unknown, although clinical reports [Shimohata et al. 2007, 2008; Glass et al. 2006; Cormican et al. 2004], experimental [Tsuda et al. 2002] and neuropathological evidence [Tada et al. 2009; Benarroch, 2007] suggest that respiratory dysfunction may be the leading cause. Future preclinical studies should help to determine the underlying mechanisms before testing specific treatment strategies.
Considerations for future clinical trials
MSA is a rare neurodegenerative disease, with an estimated prevalence ranging from 1.9 [Tison et al. 2000] to 5 cases per 100,000 persons [Vanacore et al. 2001; Schrag et al. 1999]. The long-term follow up is limited by the rapid neurodegenerative process leading to reduced life expectancy. Rapid progression may also explain the high rate of drop out in MSA trials [Dodel et al. 2010]. Therefore, prospective trials with large sample sizes are challenging. The development of multicentric network trials in the framework of the European Multiple System Atrophy-Study Group (EMSA-SG) [Geser et al. 2005] or the North American Multiple System Atrophy Study Group (NAMSAS-SG) in the United States [Gilman et al. 2005] may help to overcome this problem. For a better comparison of outcome results, clinical trials should apply consensus criteria for the diagnosis of MSA [Gilman et al. 2008] and assess disease severity by the validated UMSARS [Wenning et al. 2004].
As in other neurodegenerative disorders, a putative neuroprotective drug should be given as early as possible. Therefore, sensitivity and specificity for the diagnosis of MSA have to be improved in early disease stages. In an ideal world, patients at risk of developing MSA should be identified before the onset of core features. However, even when using recent consensual criteria [Gilman et al. 2008] current diagnostic accuracy at the first neurological evaluation remains poor [Osaki et al. 2009]. Early autonomic symptoms or preceding RBD may be useful for early MSA detection [Gaig et al. 2008; Plazzi et al. 1997; Tison et al. 1995], but whether a patient with RBD will develop MSA, PD or no neurodegenerative disorder remains unpredictable [Schenck et al. 1996].
More sensitive progression biomarkers are also needed to assess the small effect of a putative neuroprotective drug in trials. To date, cerebrospinal fluid or blood biomarkers, even in combination, are still not specific enough or need further evidence-based studies before they can be employed in routine use for the clinical diagnosis of MSA [Constantinescu et al. 2010; Mollenhauer and Trenkwalder, 2009]. Promising biomarkers may consist of structural and functional neuroimaging abnormalities [Brooks and Seppi, 2009]. While routine MRI techniques are not sensitive enough to assess disease progression in clinical trials, the use of diffusion-weighted imaging, voxel-based morphometry or scales that rate the progression of brain atrophy may be better quantitative markers for the assessment of disease-modifying properties of a neuroprotective drug [Kollensperger and Wenning, 2009].
Conclusion
Current symptomatic management in MSA should target motor impairment, autonomic failure and depression, as these features are associated with a poor quality of life [Schrag et al. 2006]. Owing to the low number of randomized and controlled trials, practical management is currently based on empirical evidence. Levodopa remains the main treatment for MSA, despite its modest and nonsustained effect. Among the several treatments available for OH, only midodrine meets the criteria of evidence-based medicine [Wright et al. 1998; Low et al. 1997; Jankovic et al. 1993]. Strategies for urinary disorders are well standardized, while other symptoms such as breathing disorders, RBD, depression or dystonia remain out of consensus.
Although there have been major advances in our understanding of the cellular pathology and in performing prospective trials designed for putative neuroprotective drugs, no curative treatment is yet available. The results from clinical trials assessing the disease-modifying potential of rasagiline and lithium will soon be available. Recent advances in genetic models and neurotransplantation may stimulate further clinical trials in MSA. Finally, the growing number of multicentric networks in Europe and North America combining preclinical and clinical research strategies provide some hope for future advances.
Conflict of interest statement
The authors declare that there are no conflicts of interest.
References
- Anderson K.N., Shneerson J.M. (2009) Drug treatment of REM sleep behavior disorder: the use of drug therapies other than clonazepam. J Clin Sleep Med 5: 235–239 [PMC free article] [PubMed] [Google Scholar]
- Apostolidis A., Dasgupta P., Denys P., Elneil S., Fowler C.J., Giannantoni A., et al. (2009) Recommendations on the use of botulinum toxin in the treatment of lower urinary tract disorders and pelvic floor dysfunctions: a European consensus report. Eur Urol 55: 100–119 [DOI] [PubMed] [Google Scholar]
- Azher S.N., Jankovic J. (2005) Camptocormia: pathogenesis, classification, and response to therapy. Neurology 65: 355–359 [DOI] [PubMed] [Google Scholar]
- Bannister R., Gibson W., Michaels L., Oppenheimer D.R. (1981) Laryngeal abductor paralysis in multiple system atrophy. A report on three necropsied cases, with observations on the laryngeal muscles and the nuclei ambigui. Brain 104: 351–368 [DOI] [PubMed] [Google Scholar]
- Beaulieu J.M., Caron M.G. (2008) Looking at lithium: molecular moods and complex behaviour. Mol Interv 8: 230–241 [DOI] [PubMed] [Google Scholar]
- Bedlack R.S., Maragakis N., Heiman-Patterson T. (2008) Lithium may slow progression of amyotrophic lateral sclerosis, but further study is needed. Proc Natl Acad Sci U S A 105: E17–E17 author reply E18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benarroch E.E. (2007) Brainstem respiratory control: substrates of respiratory failure of multiple system atrophy. Mov Disord 22: 155–161 [DOI] [PubMed] [Google Scholar]
- Bensimon G., Ludolph A., Agid Y., Vidailhet M., Payan C., Leigh P.N. (2009) Riluzole treatment, survival and diagnostic criteria in Parkinson plus disorders: the NNIPPS study. Brain 132: 156–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bloch F., Houeto J.L., Tezenas du Montcel S., Bonneville F., Etchepare F., Welter M.L., et al. (2006) Parkinson’s disease with camptocormia. J Neurol Neurosurg Psychiatry 77: 1223–1228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boesch S.M., Wenning G.K., Ransmayr G., Poewe W. (2002) Dystonia in multiple system atrophy. J Neurol Neurosurg Psychiatry 72: 300–303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boeve B.F., Silber M.H., Ferman T.J. (2003) Melatonin for treatment of REM sleep behavior disorder in neurologic disorders: results in 14 patients. Sleep Med 4: 281–284 [DOI] [PubMed] [Google Scholar]
- Brooks D.J., Seppi K. (2009) Proposed neuroimaging criteria for the diagnosis of multiple system atrophy. Mov Disord 24: 949–964 [DOI] [PubMed] [Google Scholar]
- Chen H., Jacobs E., Schwarzschild M.A., McCullough M.L., Calle E.E., Thun M.J., et al. (2005) Nonsteroidal antiinflammatory drug use and the risk for Parkinson’s disease. Ann Neurol 58: 963–967 [DOI] [PubMed] [Google Scholar]
- Chokroverty S., Sharp J.T., Barron K.D. (1978) Periodic respiration in erect posture in Shy-Drager syndrome. J Neurol Neurosurg Psychiatry 41: 980–986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colosimo C., Albanese A., Hughes A.J., de Bruin V.M., Lees A.J. (1995) Some specific clinical features differentiate multiple system atrophy (striatonigral variety) from Parkinson’s disease. Arch Neurol 52: 294–298 [DOI] [PubMed] [Google Scholar]
- Constantinescu R., Richard I., Kurlan R. (2007) Levodopa responsiveness in disorders with Parkinsonism: a review of the literature. Mov Disord 22: 2141–2148 quiz 2295. [DOI] [PubMed] [Google Scholar]
- Constantinescu R., Rosengren L., Johnels B., Zetterberg H., Holmberg B. (2010) Consecutive analyses of cerebrospinal fluid axonal and glial markers in Parkinson’s disease and atypical Parkinsonian disorders. Parkinsonism Relat Disord 16: 142–145 [DOI] [PubMed] [Google Scholar]
- Cormican L.J., Higgins S., Davidson A.C., Howard R., Williams A.J. (2004) Multiple system atrophy presenting as central sleep apnoea. Eur Respir J 24: 323–325 [DOI] [PubMed] [Google Scholar]
- de Seze M.P., Creuze A., de Seze M., Mazaux J.M. (2008) An orthosis and physiotherapy programme for camptocormia: a prospective case study. J Rehabil Med 40: 761–765 [DOI] [PubMed] [Google Scholar]
- Deguchi K., Ikeda K., Sasaki I., Shimamura M., Urai Y., Tsukaguchi M., et al. (2007) Effects of daily water drinking on orthostatic and postprandial hypotension in patients with multiple system atrophy. J Neurol 254: 735–740 [DOI] [PubMed] [Google Scholar]
- Diederich N.J., Goebel H.H., Dooms G., Bumb A., Huber F., Kompoliti K., et al. (2006) Camptocormia associated with focal myositis in multiple-system atrophy. Mov Disord 21: 390–394 [DOI] [PubMed] [Google Scholar]
- Diguet E., Fernagut P.O., Scherfler C., Wenning G., Tison F. (2005) Effects of riluzole on combined MPTP + 3-nitropropionic acid-induced mild to moderate striatonigral degeneration in mice. J Neural Transm 112: 613–631 [DOI] [PubMed] [Google Scholar]
- Dodel R., Spottke A., Gerhard A., Reuss A., Reinecker S., Schimke N., et al. (2010) Minocycline 1-year therapy in multiple-system-atrophy: effect on clinical symptoms and [(11)C] (R)-PK11195 PET (MEMSA-trial). Mov Disord 25: 97–107 [DOI] [PubMed] [Google Scholar]
- Eichhorn T.E., Oertel W.H. (2001) Macrogol 3350/electrolyte improves constipation in Parkinson’s disease and multiple system atrophy. Mov Disord 16: 1176–1177 [DOI] [PubMed] [Google Scholar]
- Fearnley J.M., Lees A.J. (1990) Striatonigral degeneration. A clinicopathological study. Brain 113: 1823–1842 [DOI] [PubMed] [Google Scholar]
- Feng H.L., Leng Y., Ma C.H., Zhang J., Ren M., Chuang D.M. (2008) Combined lithium and valproate treatment delays disease onset, reduces neurological deficits and prolongs survival in an amyotrophic lateral sclerosis mouse model. Neuroscience 155: 567–572 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernagut P.O., Ghorayeb I., Diguet E., Tison F. (2005) In vivo models of multiple system atrophy. Mov Disord 20(Suppl 12): S57–S63 [DOI] [PubMed] [Google Scholar]
- Ferrucci M., Spalloni A., Bartalucci A., Cantafora E., Fulceri F., Nutini M., et al. (2010) A systematic study of brainstem motor nuclei in a mouse model of ALS, the effects of lithium. Neurobiol Dis 37: 370–383 [DOI] [PubMed] [Google Scholar]
- Fetoni V., Soliveri P., Monza D., Testa D., Girotti F. (1999) Affective symptoms in multiple system atrophy and Parkinson’s disease: response to levodopa therapy. J Neurol Neurosurg Psychiatry 66: 541–544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fornai F., Longone P., Cafaro L., Kastsiuchenka O., Ferrucci M., Manca M.L., et al. (2008) Lithium delays progression of amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 105: 2052–2057 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fowler C.J., O'Malley K.J. (2003) Investigation and management of neurogenic bladder dysfunction. J Neurol Neurosurg Psychiatry 74(Suppl 4): iv27–iv31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freed C.R., Greene P.E., Breeze R.E., Tsai W.Y., DuMouchel W., Kao R., et al. (2001) Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med 344: 710–719 [DOI] [PubMed] [Google Scholar]
- Freeman R. (2008) Clinical practice. Neurogenic orthostatic hypotension. N Engl J Med 358: 615–624 [DOI] [PubMed] [Google Scholar]
- Freeman R., Landsberg L., Young J. (1999) The treatment of neurogenic orthostatic hypotension with 3,4-DL-threo-dihydroxyphenylserine: a randomized, placebo-controlled, crossover trial. Neurology 53: 2151–2157 [DOI] [PubMed] [Google Scholar]
- Freeman T.B., Olanow C.W., Hauser R.A., Nauert G.M., Smith D.A., Borlongan C.V., et al. (1995) Bilateral fetal nigral transplantation into the postcommissural putamen in Parkinson’s disease. Ann Neurol 38: 379–388 [DOI] [PubMed] [Google Scholar]
- Fregni F., Santos C.M., Myczkowski M.L., Rigolino R., Gallucci-Neto J., Barbosa E.R., et al. (2004) Repetitive transcranial magnetic stimulation is as effective as fluoxetine in the treatment of depression in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry 75: 1171–1174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Friess E., Kuempfel T., Modell S., Winkelmann J., Holsboer F., Ising M., et al. (2006) Paroxetine treatment improves motor symptoms in patients with multiple system atrophy. Parkinsonism Relat Disord 12: 432–437 [DOI] [PubMed] [Google Scholar]
- Gaig C., Iranzo A., Tolosa E., Vilaseca I., Rey M.J., Santamaria J. (2008) Pathological description of a non-motor variant of multiple system atrophy. J Neurol Neurosurg Psychiatry 79: 1399–1400 [DOI] [PubMed] [Google Scholar]
- Gazulla J., Benavente M.I. (2005) [Improvements in the symptoms of olivopontocerebellar atrophy with gabapentin]. Rev Neurol 40: 285–288 [PubMed] [Google Scholar]
- Gerton B.K., Theeler B., Samii A. (2010) Backpack treatment for camptocormia. Mov Disord 25: 247–248 [DOI] [PubMed] [Google Scholar]
- Geser F., Seppi K., Stampfer-Kountchev M., Kollensperger M., Diem A., Ndayisaba J.P., et al. (2005) The European Multiple System Atrophy-Study Group (EMSA-SG). J Neural Transm 112: 1677–1686 [DOI] [PubMed] [Google Scholar]
- Ghorayeb I., Bioulac B., Tison F. (2005a) Sleep disorders in multiple system atrophy. J Neural Transm 112: 1669–1675 [DOI] [PubMed] [Google Scholar]
- Ghorayeb I., Yekhlef F., Bioulac B., Tison F. (2005b) Continuous positive airway pressure for sleep-related breathing disorders in multiple system atrophy: long-term acceptance. Sleep Med 6: 359–362 [DOI] [PubMed] [Google Scholar]
- Giannantoni A., Rossi A., Mearini E., Del Zingaro M., Porena M., Berardelli A. (2009) Botulinum toxin A for overactive bladder and detrusor muscle overactivity in patients with Parkinson’s disease and multiple system atrophy. J Urol 182: 1453–1457 [DOI] [PubMed] [Google Scholar]
- Gilman S., May S.J., Shults C.W., Tanner C.M., Kukull W., Lee V.M., et al. (2005) The North American Multiple System Atrophy Study Group. J Neural Transm 112: 1687–1694 [DOI] [PubMed] [Google Scholar]
- Gilman S., Wenning G.K., Low P.A., Brooks D.J., Mathias C.J., Trojanowski J.Q., et al. (2008) Second consensus statement on the diagnosis of multiple system atrophy. Neurology 71: 670–676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glass G.A., Josephs K.A., Ahlskog J.E. (2006) Respiratory insufficiency as the primary presenting symptom of multiple-system atrophy. Arch Neurol 63: 978–981 [DOI] [PubMed] [Google Scholar]
- Hardy J. (2008) Multiple system atrophy: pathophysiology, treatment and nursing care. Nurs Stand 22: 50–56 quiz 58. [DOI] [PubMed] [Google Scholar]
- Hauser R.A., Freeman T.B., Snow B.J., Nauert M., Gauger L., Kordower J.H., et al. (1999) Long-term evaluation of bilateral fetal nigral transplantation in Parkinson disease. Arch Neurol 56: 179–187 [DOI] [PubMed] [Google Scholar]
- Hayashi M., Isozaki E., Oda M., Tanabe H., Kimura J. (1997) Loss of large myelinated nerve fibres of the recurrent laryngeal nerve in patients with multiple system atrophy and vocal cord palsy. J Neurol Neurosurg Psychiatry 62: 234–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heo J.H., Lee S.T., Chu K., Kim M. (2008) The efficacy of combined estrogen and buspirone treatment in olivopontocerebellar atrophy. J Neurol Sci 271: 87–90 [DOI] [PubMed] [Google Scholar]
- Hirohata M., Ono K., Morinaga A., Yamada M. (2008) Non-steroidal anti-inflammatory drugs have potent anti-fibrillogenic and fibril-destabilizing effects for alpha-synuclein fibrils in vitro. Neuropharmacology 54: 620–627 [DOI] [PubMed] [Google Scholar]
- Hirsch E.C., Hunot S. (2009) Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol 8: 382–397 [DOI] [PubMed] [Google Scholar]
- Holmberg B., Johansson J.O., Poewe W., Wenning G., Quinn N.P., Mathias C., et al. (2007) Safety and tolerability of growth hormone therapy in multiple system atrophy: a double-blind, placebo-controlled study. Mov Disord 22: 1138–1144 [DOI] [PubMed] [Google Scholar]
- Hughes A.J., Colosimo C., Kleedorfer B., Daniel S.E., Lees A.J. (1992) The dopaminergic response in multiple system atrophy. J Neurol Neurosurg Psychiatry 55: 1009–1013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hussain I.F., Brady C.M., Swinn M.J., Mathias C.J., Fowler C.J. (2001) Treatment of erectile dysfunction with sildenafil citrate (Viagra) in Parkinsonism due to Parkinson’s disease or multiple system atrophy with observations on orthostatic hypotension. J Neurol Neurosurg Psychiatry 71: 371–374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iranzo A. (2005) Management of sleep-disordered breathing in multiple system atrophy. Sleep Med 6: 297–300 [DOI] [PubMed] [Google Scholar]
- Iranzo A., Santamaria J., Tolosa E. (2000) Continuous positive air pressure eliminates nocturnal stridor in multiple system atrophy. Barcelona Multiple System Atrophy Study Group. Lancet 356: 1329–1330 [DOI] [PubMed] [Google Scholar]
- Iranzo A., Santamaria J., Tolosa E., Vilaseca I., Valldeoriola F., Marti M.J., et al. (2004) Long-term effect of CPAP in the treatment of nocturnal stridor in multiple system atrophy. Neurology 63: 930–932 [DOI] [PubMed] [Google Scholar]
- Ito T., Sakakibara R., Yasuda K., Yamamoto T., Uchiyama T., Liu Z., et al. (2006) Incomplete emptying and urinary retention in multiple-system atrophy: when does it occur and how do we manage it? Mov Disord 21: 816–823 [DOI] [PubMed] [Google Scholar]
- Jain S., Dawson J., Quinn N.P., Playford E.D. (2004) Occupational therapy in multiple system atrophy: a pilot randomized controlled trial. Mov Disord 19: 1360–1364 [DOI] [PubMed] [Google Scholar]
- Jankovic J., Gilden J.L., Hiner B.C., Kaufmann H., Brown D.C., Coghlan C.H., et al. (1993) Neurogenic orthostatic hypotension: a double-blind, placebo-controlled study with midodrine. Am J Med 95: 38–48 [DOI] [PubMed] [Google Scholar]
- Jellinger K.A., Lantos P.L. (2010) Papp-Lantos inclusions and the pathogenesis of multiple system atrophy: an update. Acta Neuropathol 119: 657–667 [DOI] [PubMed] [Google Scholar]
- Jin K., Okabe S., Chida K., Abe N., Kimpara T., Ohnuma A., et al. (2007) Tracheostomy can fatally exacerbate sleep-disordered breathing in multiple system atrophy. Neurology 68: 1618–1621 [DOI] [PubMed] [Google Scholar]
- Kahle P.J., Neumann M., Ozmen L., Muller V., Jacobsen H., Spooren W., et al. (2002) Hyperphosphorylation and insolubility of alpha-synuclein in transgenic mouse oligodendrocytes. EMBO Rep 3: 583–588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaufmann H. (2008) L-dihydroxyphenylserine (Droxidopa): a new therapy for neurogenic orthostatic hypotension: the US experience. Clin Auton Res 18(Suppl 1): 19–24 [DOI] [PubMed] [Google Scholar]
- Kaufmann H., Saadia D., Voustianiouk A., Goldstein D.S., Holmes C., Yahr M.D., et al. (2003) Norepinephrine precursor therapy in neurogenic orthostatic hypotension. Circulation 108: 724–728 [DOI] [PubMed] [Google Scholar]
- Kawai Y., Suenaga M., Takeda A., Ito M., Watanabe H., Tanaka F., et al. (2008) Cognitive impairments in multiple system atrophy: MSA-C vs MSA-P. Neurology 70: 1390–1396 [DOI] [PubMed] [Google Scholar]
- Kitayama M., Wada-Isoe K., Irizawa Y., Nakashima K. (2009) Assessment of dementia in patients with multiple system atrophy. Eur J Neurol 16: 589–594 [DOI] [PubMed] [Google Scholar]
- Klos K.J., Bower J.H., Josephs K.A., Matsumoto J.Y., Ahlskog J.E. (2005) Pathological hypersexuality predominantly linked to adjuvant dopamine agonist therapy in Parkinson’s disease and multiple system atrophy. Parkinsonism Relat Disord 11: 381–386 [DOI] [PubMed] [Google Scholar]
- Kollensperger M., Geser F., Seppi K., Stampfer-Kountchev M., Sawires M., Scherfler C., et al. (2008) Red flags for multiple system atrophy. Mov Disord 23: 1093–1099 [DOI] [PubMed] [Google Scholar]
- Kollensperger M., Stefanova N., Pallua A., Puschban Z., Dechant G., Hainzer M., et al. (2009) Striatal transplantation in a rodent model of multiple system atrophy: effects on L-Dopa response. J Neurosci Res 87: 1679–1685 [DOI] [PubMed] [Google Scholar]
- Kollensperger M., Wenning G.K. (2009) Assessing disease progression with MRI in atypical parkinsonian disorders. Mov Disord 24(Suppl 2): S699–S702 [DOI] [PubMed] [Google Scholar]
- Kordower J.H., Chu Y., Hauser R.A., Freeman T.B., Olanow C.W. (2008a) Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat Med 14: 504–506 [DOI] [PubMed] [Google Scholar]
- Kordower J.H., Chu Y., Hauser R.A., Olanow C.W., Freeman T.B. (2008b) Transplanted dopaminergic neurons develop PD pathologic changes: a second case report. Mov Disord 23: 2303–2306 [DOI] [PubMed] [Google Scholar]
- Kordower J.H., Freeman T.B., Chen E.Y., Mufson E.J., Sanberg P.R., Hauser R.A., et al. (1998) Fetal nigral grafts survive and mediate clinical benefit in a patient with Parkinson’s disease. Mov Disord 13: 383–393 [DOI] [PubMed] [Google Scholar]
- Lambrecq V., Krim E., Meissner W., Guehl D., Tison F. (2008) [Deep-brain stimulation of the internal pallidum in multiple system atrophy]. Rev Neurol (Paris) 164: 398–402 [DOI] [PubMed] [Google Scholar]
- Landers M., Adams M., Acosta K., Fox A. (2009) Challenge-oriented gait and balance training in sporadic olivopontocerebellar atrophy: a case study. J Neurol Phys Ther 33: 160–168 [DOI] [PubMed] [Google Scholar]
- Lee P.H., Kim J.W., Bang O.Y., Ahn Y.H., Joo I.S., Huh K. (2008) Autologous mesenchymal stem cell therapy delays the progression of neurological deficits in patients with multiple system atrophy. Clin Pharmacol Ther 83: 723–730 [DOI] [PubMed] [Google Scholar]
- Lindvall O., Brundin P., Widner H., Rehncrona S., Gustavii B., Frackowiak R., et al. (1990) Grafts of fetal dopamine neurons survive and improve motor function in Parkinson’s disease. Science 247: 574–577 [DOI] [PubMed] [Google Scholar]
- Lindvall O., Sawle G., Widner H., Rothwell J.C., Bjorklund A., Brooks D., et al. (1994) Evidence for long-term survival and function of dopaminergic grafts in progressive Parkinson’s disease. Ann Neurol 35: 172–180 [DOI] [PubMed] [Google Scholar]
- Lipp A., Sandroni P., Ahlskog J.E., Fealey R.D., Kimpinski K., Iodice V., et al. (2009) Prospective differentiation of multiple system atrophy from Parkinson disease, with and without autonomic failure. Arch Neurol 66: 742–750 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Z., Sakakibara R., Odaka T., Uchiyama T., Yamamoto T., Ito T., et al. (2005) Mosapride citrate, a novel 5-HT4 agonist and partial 5-HT3 antagonist, ameliorates constipation in parkinsonian patients. Mov Disord 20: 680–686 [DOI] [PubMed] [Google Scholar]
- Low P.A., Gilden J.L., Freeman R., Sheng K.N., McElligott M.A. (1997) Efficacy of midodrine vs placebo in neurogenic orthostatic hypotension. A randomized, double-blind multicenter study. Midodrine Study Group. JAMA 277: 1046–1051 [PubMed] [Google Scholar]
- Mancini F., Zangaglia R., Cristina S., Sommaruga M.G., Martignoni E., Nappi G., et al. (2003) Double-blind, placebo-controlled study to evaluate the efficacy and safety of botulinum toxin type A in the treatment of drooling in Parkinsonism. Mov Disord 18: 685–688 [DOI] [PubMed] [Google Scholar]
- Manni R., Morini R., Martignoni E., Pacchetti C., Micieli G., Tartara A. (1993) Nocturnal sleep in multisystem atrophy with autonomic failure: polygraphic findings in ten patients. J Neurol 240: 249–250 [DOI] [PubMed] [Google Scholar]
- Margraf N.G., Wrede A., Rohr A., Schulz-Schaeffer W.J., Raethjen J., Eymess A., et al. (2010) Camptocormia in idiopathic Parkinson’s disease: a focal myopathy of the paravertebral muscles. Mov Disord 25: 542–551 [DOI] [PubMed] [Google Scholar]
- Mathias C.J. (2008) L-dihydroxyphenylserine (Droxidopa) in the treatment of orthostatic hypotension: the European experience. Clin Auton Res 18(Suppl 1): 25–29 [DOI] [PubMed] [Google Scholar]
- Mathias C.J., Senard J.M., Braune S., Watson L., Aragishi A., Keeling J.E., et al. (2001) L-threo-dihydroxyphenylserine (l-threo-DOPS; droxidopa) of neurogenic orthostatic hypotension: a multi-national, multi-center, dose-ranging study in multiple system atrophy and pure autonomic failure. Clin Auton Res 11: 235–242 [DOI] [PubMed] [Google Scholar]
- McClellan K.J., Wiseman L.R., Wilde M.I. (1998) Midodrine. A review of its therapeutic use in the management of orthostatic hypotension. Drugs Aging 12: 76–86 [DOI] [PubMed] [Google Scholar]
- Meissner W.G., Vital, A., Ghorayeb I., Guehl D., Tison F. (2010) Dyspnea as first sign of autonomic failure in postmortem confirmed multiple system atrophy. Mov Disord in press. [DOI] [PubMed] [Google Scholar]
- Merlo I.M., Occhini A., Pacchetti C., Alfonsi E. (2002) Not paralysis, but dystonia causes stridor in multiple system atrophy. Neurology 58: 649–652 [DOI] [PubMed] [Google Scholar]
- Mollenhauer B., Trenkwalder C. (2009) Neurochemical biomarkers in the differential diagnosis of movement disorders. Mov Disord 24: 1411–1426 [DOI] [PubMed] [Google Scholar]
- Nanri K., Okita M., Takeguchi M., Taguchi T., Ishiko T., Saito H., et al. (2009) Intravenous immunoglobulin therapy for autoantibody-positive cerebellar ataxia. Intern Med 48: 783–790 [DOI] [PubMed] [Google Scholar]
- O’Sullivan J.D. (2002) Apomorphine as an alternative to sildenafil in Parkinson’s disease. J Neurol Neurosurg Psychiatry 72: 681–681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Sullivan J.D., Hughes A.J. (1998) Apomorphine-induced penile erections in Parkinson’s disease. Mov Disord 13: 536–539 [DOI] [PubMed] [Google Scholar]
- Olanow C.W., Goetz C.G., Kordower J.H., Stoessl A.J., Sossi V., Brin M.F., et al. (2003) A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Ann Neurol 54: 403–414 [DOI] [PubMed] [Google Scholar]
- Olanow C.W., Gracies J.M., Goetz C.G., Stoessl A.J., Freeman T., Kordower J.H., et al. (2009a) Clinical pattern and risk factors for dyskinesias following fetal nigral transplantation in Parkinson’s disease: a double blind video-based analysis. Mov Disord 24: 336–343 [DOI] [PubMed] [Google Scholar]
- Olanow C.W., Rascol O., Hauser R., Feigin P.D., Jankovic J., Lang A., et al. (2009b) A double-blind, delayed-start trial of rasagiline in Parkinson’s disease. N Engl J Med 361: 1268–1278 [DOI] [PubMed] [Google Scholar]
- Ono K., Hirohata M., Yamada M. (2007) Anti-fibrillogenic and fibril-destabilizing activities of anti-Parkinsonian agents for alpha-synuclein fibrils in vitro. J Neurosci Res 85: 1547–1557 [DOI] [PubMed] [Google Scholar]
- Ono K., Yamada M. (2006) Antioxidant compounds have potent anti-fibrillogenic and fibril-destabilizing effects for alpha-synuclein fibrils in vitro. J Neurochem 97: 105–115 [DOI] [PubMed] [Google Scholar]
- Osaki Y., Ben-Shlomo Y., Lees A.J., Wenning G.K., Quinn N.P. (2009) A validation exercise on the new consensus criteria for multiple system atrophy. Mov Disord 24: 2272–2276 [DOI] [PubMed] [Google Scholar]
- Papapetropoulos S., Tuchman A., Sengun C., Russell A., Mitsi G., Singer C. (2008) Anterocollis: clinical features and treatment options. Med Sci Monit 14: CR427–CR430 [PubMed] [Google Scholar]
- Papatsoris A.G., Papapetropoulos S., Singer C., Deliveliotis C. (2008) Urinary and erectile dysfunction in multiple system atrophy (MSA). Neurourol Urodyn 27: 22–27 [DOI] [PubMed] [Google Scholar]
- Pezzoli G., Tesei S., Canesi M., Sacilotto G., Vittorio M., Mizuno Y., et al. (2010) The effect of repeated administrations of granulocyte colony stimulating factor for blood stem cells mobilization in patients with progressive supranuclear palsy, corticobasal degeneration and multiple system atrophy. Clin Neurol Neurosurg 112: 65–67 [DOI] [PubMed] [Google Scholar]
- Plazzi G., Corsini R., Provini F., Pierangeli G., Martinelli P., Montagna P., et al. (1997) REM sleep behavior disorders in multiple system atrophy. Neurology 48: 1094–1097 [DOI] [PubMed] [Google Scholar]
- Puschban Z., Stefanova N., Petersen A., Winkler C., Brundin P., Poewe W., et al. (2005) Evidence for dopaminergic re-innervation by embryonic allografts in an optimized rat model of the Parkinsonian variant of multiple system atrophy. Brain Res Bull 68: 54–58 [DOI] [PubMed] [Google Scholar]
- Quinn N., Barker R.A., Wenning G.K. (2008) Are trials of intravascular infusions of autologous mesenchymal stem cells in patients with multiple system atrophy currently justified, and are they effective? Clin Pharmacol Ther 83: 663–665 [DOI] [PubMed] [Google Scholar]
- Rajput A.H., Rozdilsky B., Rajput A., Ang L. (1990) Levodopa efficacy and pathological basis of Parkinson syndrome. Clin Neuropharmacol 13: 553–558 [DOI] [PubMed] [Google Scholar]
- Ringman J.M., Simmons J.H. (2000) Treatment of REM sleep behavior disorder with donepezil: a report of three cases. Neurology 55: 870–871 [DOI] [PubMed] [Google Scholar]
- Sakakibara R., Hattori T., Uchiyama T., Suenaga T., Takahashi H., Yamanishi T., et al. (2000) Are alpha-blockers involved in lower urinary tract dysfunction in multiple system atrophy? A comparison of prazosin and moxisylyte. J Auton Nerv Syst 79: 191–195 [DOI] [PubMed] [Google Scholar]
- Sakakibara R., Matsuda S., Uchiyama T., Yoshiyama M., Yamanishi T., Hattori T. (2003) The effect of intranasal desmopressin on nocturnal waking in urination in multiple system atrophy patients with nocturnal polyuria. Clin Auton Res 13: 106–108 [DOI] [PubMed] [Google Scholar]
- Sakakibara R., Yamaguchi T., Uchiyama T., Yamamoto T., Ito T., Liu Z., et al. (2007) Calcium polycarbophil improves constipation in primary autonomic failure and multiple system atrophy subjects. Mov Disord 22: 1672–1673 [DOI] [PubMed] [Google Scholar]
- Samii A., Etminan M., Wiens M.O., Jafari S. (2009) NSAID use and the risk of Parkinson’s disease: systematic review and meta-analysis of observational studies. Drugs Aging 26: 769–779 [DOI] [PubMed] [Google Scholar]
- Santens P., Vonck K., De Letter M., Van Driessche K., Sieben A., De Reuck J., et al. (2006) Deep brain stimulation of the internal pallidum in multiple system atrophy. Parkinsonism Relat Disord 12: 181–183 [DOI] [PubMed] [Google Scholar]
- Schenck C.H., Bundlie S.R., Mahowald M.W. (1996) Delayed emergence of a parkinsonian disorder in 38% of 29 older men initially diagnosed with idiopathic rapid eye movement sleep behaviour disorder. Neurology 46: 388–393 [DOI] [PubMed] [Google Scholar]
- Schrag A., Ben-Shlomo Y., Quinn N.P. (1999) Prevalence of progressive supranuclear palsy and multiple system atrophy: a cross-sectional study. Lancet 354: 1771–1775 [DOI] [PubMed] [Google Scholar]
- Schrag A., Geser F., Stampfer-Kountchev M., Seppi K., Sawires M., Kollensperger M., et al. (2006) Health-related quality of life in multiple system atrophy. Mov Disord 21: 809–815 [DOI] [PubMed] [Google Scholar]
- Schrag A., Wenning G.K., Quinn N., Ben-Shlomo Y. (2008) Survival in multiple system atrophy. Mov Disord 23: 294–296 [DOI] [PubMed] [Google Scholar]
- Seppi K., Peralta C., Diem-Zangerl A., Puschban Z., Mueller J., Poewe W., et al. (2006) Placebo-controlled trial of riluzole in multiple system atrophy. Eur J Neurol 13: 1146–1148 [DOI] [PubMed] [Google Scholar]
- Shih L.C., Tarsy D. (2007) Deep brain stimulation for the treatment of atypical Parkinsonism. Mov Disord 22: 2149–2155 [DOI] [PubMed] [Google Scholar]
- Shimohata T., Ozawa T., Nakayama H., Tomita M., Shinoda H., Nishizawa M. (2008) Frequency of nocturnal sudden death in patients with multiple system atrophy. J Neurol 255: 1483–1485 [DOI] [PubMed] [Google Scholar]
- Shimohata T., Shinoda H., Nakayama H., Ozawa T., Terajima K., Yoshizawa H., et al. (2007) Daytime hypoxemia, is sleep-disordered breathing, and laryngopharyngeal findings in multiple system atrophy. Arch Neurol 64: 856–861 [DOI] [PubMed] [Google Scholar]
- Shioda K., Nisijima K., Kato S. (2006) Electroconvulsive therapy for the treatment of multiple system atrophy with major depression. Gen Hosp Psychiatry 28: 81–83 [DOI] [PubMed] [Google Scholar]
- Shults C.W., Rockenstein E., Crews L., Adame A., Mante M., Larrea G., et al. (2005) Neurological and neurodegenerative alterations in a transgenic mouse model expressing human alpha-synuclein under oligodendrocyte promoter: implications for multiple system atrophy. J Neurosci 25: 10689–10699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silber M.H. (2008) Tracheostomy can fatally exacerbate sleep-disordered breathing in multiple system atrophy. Neurology 70: 980–980 author reply 981–982. [DOI] [PubMed] [Google Scholar]
- Silber M.H., Levine S. (2000) Stridor and death in multiple system atrophy. Mov Disord 15: 699–704 [DOI] [PubMed] [Google Scholar]
- Singer W., Sandroni P., Opfer-Gehrking T.L., Suarez G.A., Klein C.M., Hines S., et al. (2006) Pyridostigmine treatment trial in neurogenic orthostatic hypotension. Arch Neurol 63: 513–518 [DOI] [PubMed] [Google Scholar]
- Slawek J., Derejko M., Lass P., Dubaniewicz M. (2006) Camptocormia or Pisa syndrome in multiple system atrophy. Clin Neurol Neurosurg 108: 699–704 [DOI] [PubMed] [Google Scholar]
- Song I.U., Kim J.S., Lee K.S. (2008) Dopa-responsive camptocormia in a patient with multiple system atrophy. Parkinsonism Relat Disord 14: 161–163 [DOI] [PubMed] [Google Scholar]
- Stefanova N., Bucke P., Duerr S., Wenning G.K. (2009) Multiple system atrophy: an update. Lancet Neurol 8: 1172–1178 [DOI] [PubMed] [Google Scholar]
- Stefanova N., Kollensperger M., Hainzer M., Cenci A., Poewe W., Wenning G.K. (2007) High dose levodopa therapy is not toxic in multiple system atrophy: experimental evidence. Mov Disord 22: 969–973 [DOI] [PubMed] [Google Scholar]
- Stefanova N., Poewe W., Wenning G.K. (2008) Rasagiline is neuroprotective in a transgenic model of multiple system atrophy. Exp Neurol 210: 421–427 [DOI] [PubMed] [Google Scholar]
- Stefanova N., Tison F., Reindl M., Poewe W., Wenning G.K. (2005) Animal models of multiple system atrophy. Trends Neurosci 28: 501–506 [DOI] [PubMed] [Google Scholar]
- Suzuki M., Saigusa H., Shibasaki K., Kodera K. (2010) Multiple system atrophy manifesting as complex sleep-disordered breathing. Auris Nasus Larynx 37: 110–113 [DOI] [PubMed] [Google Scholar]
- Tada M., Kakita A., Toyoshima Y., Onodera O., Ozawa T., Morita T., et al. (2009) Depletion of medullary serotonergic neurons in patients with multiple system atrophy who succumbed to sudden death. Brain 132: 1810–1819 [DOI] [PubMed] [Google Scholar]
- Takei A., Hamada S., Homma S., Hamada K., Tashiro K., Hamada T. (2009) Amelioration of subacute camptocormia in multiple system atrophy by protirelin tartrate. Mov Disord 24: 2022–2023 [DOI] [PubMed] [Google Scholar]
- Talmant V., Esposito P., Stilhart B., Mohr M., Tranchant C. (2006) [Subthalamic stimulation in a patient with multiple system atrophy: a clinicopathological report]. Rev Neurol (Paris) 162: 363–370 [DOI] [PubMed] [Google Scholar]
- Tarsy D., Apetauerova D., Ryan P., Norregaard T. (2003) Adverse effects of subthalamic nucleus DBS in a patient with multiple system atrophy. Neurology 61: 247–249 [DOI] [PubMed] [Google Scholar]
- Tison F., Wenning G.K., Quinn N.P., Smith S.J. (1995) REM sleep behaviour disorder as the presenting symptom of multiple system atrophy. J Neurol Neurosurg Psychiatry 58: 379–380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tison F., Yekhlef F., Chrysostome V. (2006) Depression and self-reported depressive symptoms in multiple system atrophy compared to Parkinson’s disease. Mov Disord 21: 1056–1057 [DOI] [PubMed] [Google Scholar]
- Tison F., Yekhlef F., Chrysostome V., Balestre E., Quinn N.P., Poewe W., et al. (2002) Parkinsonism in multiple system atrophy: natural history, severity (UPDRS-III), and disability assessment compared with Parkinson’s disease. Mov Disord 17: 701–709 [DOI] [PubMed] [Google Scholar]
- Tison F., Yekhlef F., Chrysostome V., Sourgen C. (2000) Prevalence of multiple system atrophy. Lancet 355: 495–496 [DOI] [PubMed] [Google Scholar]
- Tsuda T., Onodera H., Okabe S., Kikuchi Y., Itoyama Y. (2002) Impaired chemosensitivity to hypoxia is a marker of multiple system atrophy. Ann Neurol 52: 367–371 [DOI] [PubMed] [Google Scholar]
- Ubhi K., Rockenstein E., Mante M., Inglis C., Adame A., Patrick C., et al. (2010) Alpha-synuclein deficient mice are resistant to toxin-induced multiple system atrophy. Neuroreport in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ubhi K., Rockenstein E., Mante M., Patrick C., Adame A., Thukral M., et al. (2008) Rifampicin reduces alpha-synuclein in a transgenic mouse model of multiple system atrophy. Neuroreport 19: 1271–1276 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vanacore N., Bonifati V., Fabbrini G., Colosimo C., De Michele G., Marconi R., et al. (2001) Epidemiology of multiple system atrophy. ESGAP Consortium. European Study Group on Atypical Parkinsonisms. Neurol Sci 22: 97–99 [DOI] [PubMed] [Google Scholar]
- Vanacore N., Galeotti F. (2008) A clinical specification for a randomized clinical trial on lithium in amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 105: E35–E35 author reply E36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venglar M. (2005) Case report: Tai Chi and Parkinsonism. Physiother Res Int 10: 116–121 [DOI] [PubMed] [Google Scholar]
- Vetrugno R., Liguori R., Cortelli P., Plazzi G., Vicini C., Campanini A., et al. (2007) Sleep-related stridor due to dystonic vocal cord motion and neurogenic tachypnea/tachycardia in multiple system atrophy. Mov Disord 22: 673–678 [DOI] [PubMed] [Google Scholar]
- Vetrugno R., Provini F., Cortelli P., Plazzi G., Lotti E.M., Pierangeli G., et al. (2004) Sleep disorders in multiple system atrophy: a correlative video-polysomnographic study. Sleep Med 5: 21–30 [DOI] [PubMed] [Google Scholar]
- Visser-Vandewalle V., Temel Y., Colle H., van der Linden C. (2003) Bilateral high-frequency stimulation of the subthalamic nucleus in patients with multiple system atrophy—Parkinsonism. Report of four cases. J Neurosurg 98: 882–887 [DOI] [PubMed] [Google Scholar]
- Watanabe H., Saito Y., Terao S., Ando T., Kachi T., Mukai E., et al. (2002) Progression and prognosis in multiple system atrophy: an analysis of 230 Japanese patients. Brain 125: 1070–1083 [DOI] [PubMed] [Google Scholar]
- Waxman E.A., Giasson B.I. (2010) A novel, high-efficiency cellular model of fibrillar alpha-synuclein inclusions and the examination of mutations that inhibit amyloid formation. J Neurochem 113: 374–388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wedge F. (2008) The impact of resistance training on balance and functional ability of a patient with multiple system atrophy. J Geriatr Phys Ther 31: 79–83 [DOI] [PubMed] [Google Scholar]
- Wenning G.K. (2005) Placebo-controlled trial of amantadine in multiple-system atrophy. Clin Neuropharmacol 28: 225–227 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Ben-Shlomo Y., Hughes A., Daniel S.E., Lees A., Quinn N.P. (2000) What clinical features are most useful to distinguish definite multiple system atrophy from Parkinson’s disease? J Neurol Neurosurg Psychiatry 68: 434–440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wenning G.K., Ben-Shlomo Y., Magalhaes M., Daniel S.E., Quinn N.P. (1995) Clinicopathological study of 35 cases of multiple system atrophy. J Neurol Neurosurg Psychiatry 58: 160–166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wenning G.K., Ben Shlomo Y., Magalhaes M., Daniel S.E., Quinn N.P. (1994) Clinical features and natural history of multiple system atrophy. An analysis of 100 cases. Brain 117: 835–845 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Geser F., Stampfer-Kountchev M., Tison F. (2003) Multiple system atrophy: an update. Mov Disord 18(Suppl 6): S34–S42 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Stefanova N. (2009) Recent developments in multiple system atrophy. J Neurol 256: 1791–1808 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Stefanova N., Jellinger K.A., Poewe W., Schlossmacher M.G. (2008) Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol 64: 239–246 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Tison F., Ben Shlomo Y., Daniel S.E., Quinn N.P. (1997) Multiple system atrophy: a review of 203 pathologically proven cases. Mov Disord 12: 133–147 [DOI] [PubMed] [Google Scholar]
- Wenning G.K., Tison F., Seppi K., Sampaio C., Diem A., Yekhlef F., et al. (2004) Development and validation of the Unified Multiple System Atrophy Rating Scale (UMSARS). Mov Disord 19: 1391–1402 [DOI] [PubMed] [Google Scholar]
- Whone A.L., Scolding N.J. (2009) Mesenchymal stem cells and neurodegenerative disease. Clin Pharmacol Ther 85: 19–20 [DOI] [PubMed] [Google Scholar]
- Widner H., Tetrud J., Rehncrona S., Snow B., Brundin P., Gustavii B., et al. (1992) Bilateral fetal mesencephalic grafting in two patients with Parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). N Engl J Med 327: 1556–1563 [DOI] [PubMed] [Google Scholar]
- Wright R.A., Kaufmann H.C., Perera R., Opfer-Gehrking T.L., McElligott M.A., Sheng K.N., et al. (1998) A double-blind, dose-response study of midodrine in neurogenic orthostatic hypotension. Neurology 51: 120–124 [DOI] [PubMed] [Google Scholar]
- Yamaguchi M., Arai K., Asahina M., Hattori T. (2003) Laryngeal stridor in multiple system atrophy. Eur Neurol 49: 154–159 [DOI] [PubMed] [Google Scholar]
- Yazawa I., Giasson B.I., Sasaki R., Zhang B., Joyce S., Uryu K., et al. (2005) Mouse model of multiple system atrophy alpha-synuclein expression in oligodendrocytes causes glial and neuronal degeneration. Neuron 45: 847–859 [DOI] [PubMed] [Google Scholar]
- Young T.M., Mathias C.J. (2004) The effects of water ingestion on orthostatic hypotension in two groups of chronic autonomic failure: multiple system atrophy and pure autonomic failure. J Neurol Neurosurg Psychiatry 75: 1737–1741 [DOI] [PMC free article] [PubMed] [Google Scholar]