Multiple System Atrophy - A Synucleinopathy with Specific Glioneuronal Degeneration

Mini Review

Austin J Clin Neurol 2015; 2(8): 1071.

Multiple System Atrophy - A Synucleinopathy with Specific Glioneuronal Degeneration

Jellinger KA*

Institute of Clinícal Neurobiology, Medical University of Vienna, Austria

*Corresponding author: Kurt A Jellinger, Institute of Clinical Neurobiology, Medical University of Vienna, Alberichgasse, A-1150 Vienna, Austria

Received: June 11, 2015; Accepted: August 10, 2015; Published: August 12, 2015

Abstract

Multiple system atrophy (MSA) is a rare, largely sporadic, adult-onset neurodegenerative disorder of uncertain etiology, clinically manifesting with Parkinsonism, cerebellar impairment, autonomic dysfunction, and pyramidal signs. The pathological process affects striatonigral, olivopontocerebellar, and autonomic nervous systems. The major clinical variants correlate to the morphologic phenotypes of striatonigral degeneration (MSA-P) and olivopontocerebellar atrophy (MSA-C). Pathologically, MSA is characterized by glial cytoplasmic inclusions (GCIs) and neuronal inclusions (NIs) containing abnormal filamentous α-synuclein that involve many areas of the nervous system. Recent advances have increased our knowledge of the molecular pathogenesis of this devastating disease; updated consensus criteria and combined fluid and imaging biomarkers have increased the diagnostic accuracy and sensitivity versus other parkinsonian syndromes considerably. The pathology of this unique proteinopathy, in addition to ectopic deposition of misformed α-synuclein in glia and neurons, its cell-to-cell spreading in a prion-like matter, oxidative stress, proteasomal and mitochondrial dysfunction, dysregulation of myelin lipids, decreased expression of neurotropic factors, neuro inflammation, and energy failure, contributes to system-specific neuro degeneration. Despite several pharmacological approaches in MSA models, addressing these pathogenic mechanisms, no disease-modifying treatment for MSA is currently available.

Keywords: Multiple system atrophy; Diagnostic criteria; Pathogenesis; α-synuclein; Prion-like seeding; Glio-neuronal degeneration; Candidate biomarkers; New therapeutic approaches

Abbreviations

αSyn: α-Synuclein; CBD: Corticobasal Degeneration; FTLD: Frontolemporal Lobe Dementia; GCIs: Glial Cytoplasmic Inclusions; LBD: Lewy Body Dementia; MSA: Multiple System Atrophy; MSA-C: MSA Cerebellar phenotype; MSA-P: MSA with Predominant Parkinsonism; NCIs: Neuronal Cytoplasmic Inclusions; NFL: Light Chain Neurofilament Protein; NIs: Neuronal Inclusions; PD: Parkinson Disease; PSP: Progressive Supra nuclear Palsy

Introduction

Multiple system atrophy (MSA) is a rare, largely spontaneous, rapidly progressing neurodegenerative disorder of uncertain etiology that is clinically characterized by a variable combination of Parkinsonism, cerebellar impairment, autonomic dysfunction and pyramidal tract signs. Its estimated main incidence is 0.6 to 0.7 cases/ 100,000 populations; the estimated point prevalence is 1.9 to 5 cases/ 100,000 increasing to 7.8/100,000 after age 40 years. The pathological process predominantly affects the striatonigral and olivopontocerebellar systems, which underlies the stratification of the heterogeneous disorder into a clinical phenotype with predominant Parkinsonism (MSA-P) and a cerebellar phenotype (MSA-C). In the Western hemisphere, MSA-P involves 70% of the patients, while in Asian populations MSA-C predominates in two-thirds of patients. MSA is a late-onset disorder (mean age at onset 56±9 years) with poor functional prognosis and a median survival from onset of 9.5 years [1,2]. Shorter symptom duration at baseline and absent L-dopa response predicted rapid UMSARS (unified MSA rating scale) progression [3]. MSA-P with slow progression and prolonged survival is an uncommon “benign” subgroup [4], while “minimal change” MSA is considered an aggressive variant [5].

Clinical Diagnosis

Recent consensus criteria differentiate possible, probable, and definite MSA, the latter confirmed by postmortem examination [6]. Red flag clinical categories had a specificity of 98.3% and a sensitivity of 84.2% [7]. Due to overlapping clinical presentations, it can be difficult to distinguish MSA from Parkinson disease (PD) in early disease, and from other atypical parkinsonian disorders, eg., progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) [8]. Prevalence of REM sleep behavior disorder in MSA is up to 88% [9].

No reliable fluid biomarkers are currently available to guide the clinical diagnosis and prognosis, although many studies suggest that combining CSF biomarkers, e.g. DJ-1, phospho-tau, light chain neurofilament protein (NFL), and Aβ-42 may be more successful in the differential diagnosis between MSA and other parkinsonian disorders [10]. Hypo intensity of the dorsolateral put amen in T2- weighted MRI due to iron deposition differentiates MSA-P from PD with high sensitivity [11-13]. There is recent evidence that functional MRI activation is abnormal in the basal ganglia, cerebellum, and cerebrum in MSA-P, and that another key distinguishing feature between MSA-P and PD is the extensive and widespread volume loss throughout the brain in MSA-P [14]. Recently described, rare cases of atypical MSA with clinical features consistent with frontotemporal lobe dementia (FTLD), have been suggested to represent a novel subtype of FTLD associated with αSyn [15].

Neuropathology

Together with PD and Lewy body dementia (LBD), MSA belongs to a group of neurodegenerative disorders - the α-synucleinopathies - which are characterized by the abnormal accumulation of α-synuclein (αSyn). The histological core features are glial cytoplasmic inclusions (GCIs, Papp-Lantos bodies) in oligodendroglia, the demonstration of which is required for the diagnosis of definite MSA [16]. αSyn, together with other proteins, is the main constituent of GCIs that also involves neurons as neuronal inclusions (NIs) [17] and other cells in wide areas of the nervous system, causing neuronal loss and demyelination [18]. Based on semi quantitative assessment of neuronal loss, gliosis and GCIs in brain regions, grading of striatonigral and olivopontocerebellar lesions into four degrees of severity [19] was confirmed by postmortem MRI [20], while others showed an overlap between striatonigral and Olivopontocerebellar atrophy (OPCA) system degeneration [21]. Recent stereological studies have demonstrated significant neuronal loss in substantial nigra, striatum and globus pallidus in MSA [22], and a widespread involvement of the neocortex, in particular the frontal cortex of MSA patients with impaired executive function [23]. Voxel-based morphometry (VBM) demonstrated significant gray matter atrophy in the MSA-P group in bilateral basal ganglia, cerebellum, frontal and temporal cortices, which were significantly correlated with cognitive dysfunction in MSA [24].

The lesions are not limited to the striatonigal and olivopontocerebellar systems but also involve many other parts of the central, peripheral and autosomal nervous system, underpinning the multisystem character of MSA [18,25]. Recent studies of skin biopsies revealed phospho-αSyn in Schwann cells [26] and in unmyelinated somatosensory dermal nerve fibers, whereas deposits in autonomic fibers are mainly found in PD [27].

Concomintant pathologies in MSA include Lewy bodies, the hallmark of PD and DLB, in 11 to 25.7%, frequently associated with cognitive impairment [17,18], rare co-occurrence of MSA and PSP and other tauopathies, while Alzheimer-related and TDP-43 pathologies occur infrequently in MSA [18,28].

Etiopathogenesis

The causes of MSA are unknown. No environmental factors have been recognized. MSA is generally considered a sporadic disease, but there are familial cases, and in some pedigrees, it has been transmitted in an autosomal dominant form or recessive inheritance pattern. Mutations of Coenzyme Q10 (COQ2), SNCA, encoding αSyn, glucocerebrosidase gene (GBA) variants and other genetic loci have been investigated, but their association is under discussion [29- 35]. A G51D SNCA mutation was reported in a British family with autosomal dominant Parkinsonism and neuropathological findings comparable with both PD and MSA [36], while MSA is not a C9orf72- related disease [37].

Although the mechanisms of αSyn triggered neurodegeneration and the pathogenesis of MSA are not fully understood, evidence from animal models and postmortem studies suggested that it is a primary oligodendrogliopathy [38]. The origin of αSyn-positive GCIs found in oligodendrocytes in MSA is enigmatic since earlier studies did not find expression of the protein in MSA oligodendroglia, which recently has been challenged [39]. Oligomeric αSyn and small fibrils are probably the most toxic forms initiating the aggregation process and subsequent cell death [40,41], and impairs maturation of primary oligodendrocyte progenitor cells [42,43]. Recent studies showed that αSyn can be transferred to grafted oligodendroglial cells from host rat brain neurons over expressing αSyn, supporting its neuron-tooligodendrocyte transfer [44], and suggest that – similar to preclinical models of PD – it is seeded through the brain in a “prion-like” manner in MSA [45,46].

The earliest stages of MSA pathogenesis is currently unknown but is likely to involve a relocation of p25α (TPPP), an oligodendrogliaspecific phosphoprotein, an important stabilizer of microtubules and myelin integrity [47], from the myelin sheaths into the oligodendroglial soma preceding the αSyn aggregation. Co expression of αSyn and p25α increases the expression of IκBα early and is dependent on aggregation and phosphorylation of αSyn. There is increased expression of IκBα and NF-κB in some oligodendrocytes containing GCIs, suggesting that both proteins are activate early in the course of MSA and their balance contributes to cellular demise [48]. These changes are followed by oligodendrocyte swelling and abnormal uptake or over expression of αSyn, which undergoes formation into insoluble oligomeres, followed by formation of GCIs [49]. Association with a significant decrease of p25α in oligodendroglia containing αSyn positive GCIs, implies that mitochondrial dysfunction can lead to secondary p25α relocation [50]. The ubiquitin-proteasomal pathway (UPS) and the autophagy-lysosomal pathway are tightly balanced and inhibition of the deubiquitinating enzyme carboxyl-terminal hydrolase L1 (UCH-L1) in oligodendrocytes results in microtubule stabilization and prevents αSyn aggregate formation by activating the autosomal pathway, thus playing a role in oligodendroglial degeneration [51].

Dysregulation of the specialized lipid metabolism involved in myelin synthesis is associated with these changes [52,53]. The decrease in lipid levels was concomitant with increased αSyn expression, indicating that levels and not distribution of myelin lipid are altered in MSA, triggering myelin instability [54]. The formation of GCIs interferes with oligodendroglial and neuronal trophic support leading to functional disorder and eventually death of these cells, and initiates neuroinflammation by activation of quiescent microglia [18]. Released misfolded αSyn into the extracellular space may be taken up by neighboring neurons to form neuronal cytoplasmic inclusions (NCIs); it is suggested to spread in a “prion-like” through functionally connected neuronal networks [55], resulting in a system-like pattern of neurodegeneration that is typical of MSA. Recent postmortem studies expanded the spectrum of neuronal pathology in MSA, describing increased frequencies of neuronal inclusions, both NIs and Lewy bodies across a wide spectrum of brain regions, not only in canonical disease-associated regions (striatum, substantia nigra), but also in many other region, suggesting a hierarchy of region-specific susceptibility [17]. Disease duration is significantly correlated with the severity of neurodegeneration, suggesting that the progression of αSyn pathology is time-dependent [18]; NIs appears earlier than previously thought. A correlation between neuronal pathology and both GCIs and NIs in the most severely affected brain regions, suggesting a linked between these phenomena has been reported [56], although the mechanisms underlying this remain to be elucidated.

In conclusion, the pathogenesis of MSA currently remains unknown. The disease has been viewed as a primary gliopathysynucleinopathy with neuronal pathology developing secondarily through mechanisms via the oligo-myelin-axon-neuron complex [38] Other authors have proposed that neuronal and glial inclusions may interact synergistically through unidentified mechanisms [57], disease progression resulting from the simultaneous degeneration of glia and myelin, due to GCIs, and aggregation of αSyn within neurons. On the other hand, MSA may be a primary neuronal disease and that the formation of GCIs results from secondary accumulation of pathologic αSyn that is neuronal in origin [58]. The influence of GCIs on the formation of NIs is unclear, but the burden of neuronal pathology appears to increase multifocally as an effect of disease duration associated with increasing overall αSyn load. Recent findings support the concept that neuronal pathology is an important if not primary component of MSA pathogenesis [17], which does not exclude the possibility of acceleration of neuronal pathology by accumulation of αSyn in glia as GCIs [38, 57]. Further research on the basic pathogenic mechanisms, the interplay of the disease process with various pathobiological changes, and the nature of possible genetic and environmental triggers that unmask its pathogenesis are needed to develop optimal animal models, and to clarify the relations between the development of pathomorphology and clinical manifestations as a basis for early diagnosis and a successful treatment of this hitherto incurable devastating disorder [59]. The advantages and limitations of MSA models and their application in preclinical target validation have been summarized critically [60].

Therapeutic Approaches in MSA

Currently, there is neither an effective neuroprotective nor a disease-modifying therapy in MSA although several pharmacological approaches have been tried in transgenic mouse or cellular models of MSA, including riluzole, rasagilin, minocycline, stem cells, etc., treatments that can halt or reverse the disease progression in humans have not yet been identified [61,62]. Symptomatic approaches include dopaminergic and anticholinergic agents, nonpharmacological treatment; options to treat orthostatic hypotension, urinary and erectile dysfunction as well as palliative care [63]. Active immunization against αSyn has been shown to ameliorate the degenerative pathology and to prevent demyelination in a mouse model of MSA [64]. Understanding the pathogenesis of MSA and the factors leading to αSyn accumulation is essential for the development of successful therapeutic options. Effective treatment may result from a multi-targeted approach addressing several pathophysiological mechanisms together and from multidisciplinary collaborative efforts to test promising new therapies in properly designed clinical trials.

References

  1. Fanciulli A, Wenning GK. Multiple-system atrophy. N Engl J Med. 2015; 372: 249-263.
  2. Low PA, Reich SG, Jankovic J, Shults CW, Stern MB, Novak P, et al. Natural history of multiple system atrophy in the USA: a prospective cohort study. Lancet Neurol. 2015; 14: 710-719.
  3. Wenning GK, Geser F, Krismer F, Seppi K, Duerr S, Boesch S, et al. The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol. 2013; 12: 264-274.
  4. Petrovic IN, Ling H, Asi Y, Ahmed Z, Kukkle PL, Hazrati LN, et al. Multiple system atrophy-parkinsonism with slow progression and prolonged survival: a diagnostic catch. Mov Disord. 2012; 27: 1186-1190.
  5. Ling H, Asi YT, Petrovic IN, Ahmed Z, Prashanth LK. Minimal change multiple system atrophy: An aggressive variant? Mov Disord. 2015; 30: 960-967.
  6. Gilman S, Wenning GK, Low PA, Brooks DJ, Mathias CJ, Trojanowski JQ, et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology. 2008; 71: 670-676.
  7. Köllensperger M, Geser F, Ndayisaba JP, Boesch S, Seppi K, Ostergaard K, et al. Presentation, diagnosis, and management of multiple system atrophy in Europe: final analysis of the European multiple system atrophy registry. Mov Disord. 2010; 25: 2604-2612.
  8. Kim HJ, Jeon BS, Jellinger KA. Diagnosis and differential diagnosis of MSA: boundary issues. J Neurol. 2015; .
  9. Palma JA, Fernandez-Cordon C, Coon EA, Low PA, Miglis MG, Jaradeh S, et al. Prevalence of REM sleep behavior disorder in multiple system atrophy: a multicenter study and meta-analysis. Clin Auton Res. 2015; 25: 69-75.
  10. Laurens B, Constantinescu R, Freeman R, Gerhard A, Jellinger K, Jeromin A, et al. Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative. Neurobiol Dis. 2015; 80: 29-41.
  11. Lee JH, Kim TH, Mun CW, Kim TH, Han YH. Progression of subcortical atrophy and iron deposition in multiple system atrophy: a comparison between clinical subtypes. J Neurol. 2015; .
  12. Ramli N, Nair SR, Ramli NM, Lim SY. Differentiating multiple-system atrophy from Parkinson's disease. Clin Radiol. 2015; 70: 555-564.
  13. Sugiyama A, Ito S, Suichi T, Sakurai T, Mukai H, Yokota H, et al. Putaminal hypointensity on T2*-weighted MR imaging is the most practically useful sign in diagnosing multiple system atrophy: A preliminary study. J Neurol Sci. 2015; 349: 174-178.
  14. Planetta PJ, Kurani AS, Shukla P, Prodoehl J, Corcos DM, Comella CL, et al. Distinct functional and macrostructural brain changes in Parkinson's disease and multiple system atrophy. Hum Brain Mapp. 2015; 36: 1165-1179.
  15. Aoki N, Boyer PJ, Lund C, Lin WL, Koga S, Ross OA, et al. Atypical multiple system atrophy is a new subtype of frontotemporal lobar degeneration: frontotemporal lobar degeneration associated with α-synuclein. Acta Neuropathol. 2015; 130: 93-105.
  16. Trojanowski JQ, Revesz T; Neuropathology Working Group on MSA. Proposed neuropathological criteria for the post mortem diagnosis of multiple system atrophy. Neuropathol Appl Neurobiol. 2007; 33: 615-620.
  17. Cykowski MD, Coon EA, Powell SZ, Jenkins SM, Benarroch EE, Low PA, et al. Expanding the spectrum of neuronal pathology in multiple system atrophy. Brain. 2015; 138: 2293-2309.
  18. Jellinger KA. Neuropathology of multiple system atrophy: new thoughts about pathogenesis. Mov Disord. 2014; 29: 1720-1741.
  19. Jellinger KA, Seppi K, Wenning GK. Grading of neuropathology in multiple system atrophy: proposal for a novel scale. Mov Disord. 2005; 20 Suppl 12: S29-36.
  20. Matsusue E, Fujii S, Kanasaki Y, Kaminou T, Ohama E, Ogawa T. Cerebellar lesions in multiple system atrophy: postmortem MR imaging-pathologic correlations. AJNR Am J Neuroradiol. 2009; 30: 1725-1730.
  21. Halliday GM, Holton JL, Revesz T, Dickson DW. Neuropathology underlying clinical variability in patients with synucleinopathies. Acta Neuropathol. 2011; 122: 187-204.
  22. Salvesen L, Ullerup BH, Sunay FB, Brudek T, Løkkegaard A, Agander TK, et al. Changes in total cell numbers of the basal ganglia in patients with multiple system atrophy - A stereological study. Neurobiol Dis. 2015; 74: 104-113.
  23. Salvesen L, Ullerup BH, Sunay FB, Brudek T, Løkkegaard A, Agander TK, et al. Changes in total cell numbers of the basal ganglia in patients with multiple system atrophy - A stereological study. Neurobiol Dis. 2015; 74: 104-113.
  24. Kim JS, Yang JJ, Lee DK, Lee JM, Youn J, Cho JW. Cognitive Impairment and Its Structural Correlates in the Parkinsonian Subtype of Multiple System Atrophy. Neurodegener Dis. 2015; .
  25. Ahmed Z, Asi YT, Sailer A, Lees AJ, Houlden H, Revesz T, et al. The neuropathology, pathophysiology and genetics of multiple system atrophy. Neuropathol Appl Neurobiol. 2012; 38: 4-24.
  26. Nakamura K, Mori F, Kon T, Tanji K, Miki Y, Tomiyama M. Filamentous aggregations of phosphorylated α-synuclein in Schwann cells (Schwann cell cytoplasmic inclusions) in multiple system atrophy. Acta Neuropathol Commun. 2015; 3: 29.
  27. Doppler K, Weis J, Karl K, Ebert S, Ebentheuer J, Trenkwalder C, et al. Distinctive Distribution of phospho-alpha-synuclein in dermal nerves in multiple system atrophy. Mov Disord. 2015; .
  28. Geser F, Malunda JA, Hurtig HI, Duda JE, Wenning GK, Gilman S, et al. TDP-43 pathology occurs infrequently in multiple system atrophy. Neuropathol Appl Neurobiol. 2011; 37: 358-365.
  29. Ogaki K, Fujioka S, Heckman MG, Rayaprolu S, Soto-Ortolaza AI, Labbé C, et al. Analysis of COQ2 gene in multiple system atrophy. Mol Neurodegener. 2014; 9: 44.
  30. Chen YP, Zhao B, Cao B, Song W, Guo X, Wei QQ, et al. Mutation scanning of the COQ2 gene in ethnic Chinese patients with multiple-system atrophy. Neurobiol Aging. 2015; 36: 1222.
  31. Heckman MG, Schottlaender L, Soto-Ortolaza AI, Diehl NN, Rayaprolu S, Ogaki K, et al. LRRK2 exonic variants and risk of multiple system atrophy. Neurology. 2014; 83: 2256-2261.
  32. Federoff M, Schottlaender LV, Houlden H, Singleton A. Multiple system atrophy: the application of genetics in understanding etiology. Clin Auton Res. 2015; 25: 19-36.
  33. Mitsui J, Matsukawa T, Sasaki H, Yabe I, Matsushima M, Dürr A, et al. Variants associated with Gaucher disease in multiple system atrophy. Ann Clin Transl Neurol. 2015; 2: 417-426.
  34. Scholz SW, Houlden H, Schulte C, Sharma M, Li A, Berg D, et al. SNCA variants are associated with increased risk for multiple system atrophy. Ann Neurol. 2009; 65: 610-614.
  35. Ross OA, Vilariño-Güell C, Wszolek ZK, Farrer MJ, Dickson DW. Reply to: SNCA variants are associated with increased risk of multiple system atrophy. Ann Neurol. 2010; 67: 414-415.
  36. Kiely AP, Asi YT, Kara E, Limousin P, Ling H, Lewis P, et al. α-Synucleinopathy associated with G51D SNCA mutation: a link between Parkinson's disease and multiple system atrophy? Acta Neuropathol. 2013; 125: 753-769.
  37. Scholz SW, Majounie E, Revesz T, Holton JL, Okun MS, Houlden H, et al. Multiple system atrophy is not caused by C9orf72 hexanucleotide repeat expansions. Neurobiol Aging. 2015; 36: 1223.
  38. Wenning GK, Stefanova N, Jellinger KA, Poewe W, Schlossmacher MG. Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol. 2008; 64: 239-246.
  39. Asi YT, Simpson JE, Heath PR, Wharton SB, Lees AJ, Revesz T, et al. Alpha-synuclein mRNA expression in oligodendrocytes in MSA. Glia. 2014; 62: 964-970.
  40. Winner B, Jappelli R, Maji SK, Desplats PA, Boyer L, Aigner S, et al. In vivo demonstration that alpha-synuclein oligomers are toxic. Proc Natl Acad Sci U S A. 2011; 108: 4194-4199.
  41. Marques O, Outeiro TF. Alpha-synuclein: from secretion to dysfunction and death. Cell Death Dis. 2012; 3: e350.
  42. Ettle B, Reiprich S, Deusser J, Schlachetzki JC, Xiang W, Prots I, et al. Intracellular alpha-synuclein affects early maturation of primary oligodendrocyte progenitor cells. Mol Cell Neurosci. 2014; 62: 68-78.
  43. May VE, Ettle B, Poehler AM, Nuber S, Ubhi K, Rockenstein E, et al. alpha-Synuclein impairs oligodendrocyte progenitor maturation in multiple system atrophy. Neurobiol Aging. 2014; 35: 2357-2368.
  44. Reyes JF, Rey NL, Bousset L, Melki R, Brundin P, Angot E. Alpha-synuclein transfers from neurons to oligodendrocytes. Glia. 2014; 62: 387-398.
  45. Watts JC, Giles K, Oehler A, Middleton L, Dexter DT, Gentleman SM, et al. Transmission of multiple system atrophy prions to transgenic mice. Proc Natl Acad Sci U S A. 2013; 110: 19555-19560.
  46. Kovacs GG, Breydo L, Green R, Kis V, Puska G, Lőrincz P, et al. Intracellular processing of disease-associated α-synuclein in the human brain suggests prion-like cell-to-cell spread. Neurobiol Dis. 2014; 69: 76-92.
  47. Ovádi J, Orosz F. An unstructured protein with destructive potential: TPPP/p25 in neurodegeneration. Bioessays. 2009; 31: 676-686.
  48. Kragh CL, Gysbers AM, Rockenstein E, Murphy K, Halliday GM, Masliah E, et al. Prodegenerative IκBα expression in oligodendroglial α-synuclein models of multiple system atrophy. Neurobiol Dis. 2014; 63: 171-183.
  49. Song YJ, Lundvig DM, Huang Y, Gai WP, Blumbergs PC, Højrup P, et al. p25alpha relocalizes in oligodendroglia from myelin to cytoplasmic inclusions in multiple system atrophy. Am J Pathol. 2007; 171: 1291-1303.
  50. Ota K, Obayashi M, Ozaki K, Ichinose S, Kakita A, Tada M, et al. Relocation of p25alpha/tubulin polymerization promoting protein from the nucleus to the perinuclear cytoplasm in the oligodendroglia of sporadic and COQ2 mutant multiple system atrophy. Acta Neuropathol Commun. 2014; 2: 136.
  51. Pukass K, Richter-Landsberg C. Inhibition of UCH-L1 in oligodendroglial cells results in microtubule stabilization and prevents alpha-synuclein aggregate formation by activating the autophagic pathway: implications for multiple system atrophy. Front Cell Neurosci. 2015; 9: 163.
  52. Bleasel JM, Wong JH, Halliday GM, Kim WS. Lipid dysfunction and pathogenesis of multiple system atrophy. Acta Neuropathol Commun. 2014; 2: 15.
  53. Wong JH, Halliday GM, Kim WS. Exploring myelin dysfunction in multiple system atrophy. Exp Neurobiol. 2014; 23: 337-344.
  54. Don AS, Hsiao JH, Bleasel JM, Couttas TA, Halliday GM, Kim WS. Altered lipid levels provide evidence for myelin dysfunction in multiple system atrophy. Acta Neuropathol Commun. 2014; 2: 150.
  55. Masuda-Suzukake M, Nonaka T, Hosokawa M, Kubo M, Shimozawa A, Akiyama H, et al. Pathological alpha-synuclein propagates through neural networks. Acta Neuropathol Commun. 2014; 2: 88.
  56. Ozawa T, Paviour D, Quinn NP, Josephs KA, Sangha H, Kilford L, et al. The spectrum of pathological involvement of the striatonigral and olivopontocerebellar systems in multiple system atrophy: clinicopathological correlations. Brain. 2004; 127: 2657-2671.
  57. Yoshida M. Multiple system atrophy: alpha-synuclein and neuronal degeneration. Neuropathology. 2007; 27: 484-493.
  58. Ubhi K, Low P, Masliah E. Multiple system atrophy: a clinical and neuropathological perspective. Trends Neurosci. 2011; 34: 581-590.
  59. Fernagut PO, Dehay B, Maillard A, Bezard E, Perez P, Pavy-Le Traon A, et al. Multiple system atrophy: a prototypical synucleinopathy for disease-modifying therapeutic strategies. Neurobiol Dis. 2014; 67: 133-139.
  60. Stefanova N, Wenning GK. Animal models of multiple system atrophy. Clin Auton Res. 2015; 25: 9-17.
  61. Palma JA, Kaufmann H. Novel therapeutic approaches in multiple system atrophy. Clin Auton Res. 2015; 25: 37-45.
  62. Poewe W, Seppi K, Fitzer-Attas CJ, Wenning GK, Gilman S, Low PA, et al. Efficacy of rasagiline in patients with the parkinsonian variant of multiple system atrophy: a randomised, placebo-controlled trial. Lancet Neurol. 2015; 14: 145-152.
  63. Granata R, Wenning GK. Multiple system atrophy. In: Colosimo C, Riley DE, Wenning GK, editors. Handbook of Atypical Parkinsonism. Cambridge: Cambridge University Press 2011; pp. 27-57.
  64. Mandler M, Valera E, Rockenstein E, Mante M, Weninger H, Patrick C, et al. Active immunization against alpha-synuclein ameliorates the degenerative pathology and prevents demyelination in a model of multiple system atrophy. Mol Neurodegener. 2015; 10: 10.

Download PDF

Citation: Jellinger KA. Multiple System Atrophy - A Synucleinopathy with Specific Glioneuronal Degeneration. Austin J Clin Neurol 2015; 2(8): 1071. ISSN : 2381-9154

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us