ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Multiple system atrophy: Epidemiology, pathology, and pathogenesis

Multiple system atrophy: Epidemiology, pathology, and pathogenesis
Literature review current through: Jan 2024.
This topic last updated: Feb 06, 2023.

INTRODUCTION — Multiple system atrophy (MSA) is a rare neurodegenerative disorder that pathologically involves the basal ganglia, brainstem, cerebellum, and spinal cord; it clinically presents with various combinations of autonomic dysfunction, parkinsonism, ataxia, and corticospinal degeneration.

MSA is one of several neurodegenerative disorders associated with alpha-synuclein aggregation, or synucleinopathies; others include Parkinson disease (PD) and dementia with Lewy bodies (DLB). MSA is also referred to as an atypical parkinsonian disorder based on distinct yet overlapping features with PD; other atypical parkinsonian disorders include progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD).

This topic will review the epidemiology, pathology, pathogenesis, and genetics of MSA. The clinical features, diagnosis, and treatment of MSA are reviewed separately. (See "Multiple system atrophy: Clinical features and diagnosis" and "Multiple system atrophy: Prognosis and treatment".)

HISTORY AND NOMENCLATURE — MSA is a unifying term that brings together a group of rare, fatal neurodegenerative syndromes that used to be referred to as olivopontocerebellar atrophy (OPCA), striatonigral degeneration, and Shy-Drager syndrome.

In 1900, Dejerine and Thomas provided the first report of sporadic OPCA, a disease that later would become a part of the spectrum of MSA [1]. Orthostatic hypotension as a manifestation of autonomic failure was described in 1925 [2], and in 1960, Shy and Drager reported patients with autonomic features of orthostatic syncope, impotence, and bladder dysfunction who went on to develop gait abnormalities, tremor, and fasciculations [3]. This disorder ultimately became known as the Shy-Drager syndrome. Also in 1960, the first cases of a predominantly akinetic and rigid parkinsonian syndrome were reported, which correlated pathologically with evidence of striatonigral degeneration [4].

In 1969, the term "multiple system atrophy" was introduced to encompass all three clinical syndromes, based on their similar pathology [5]. Striatonigral degeneration was later defined as MSA with predominant parkinsonism (MSA-P), while OPCA was defined as MSA with predominant cerebellar ataxia (MSA-C) [6]. Ultimately, glial cytoplasmic inclusions containing alpha-synuclein were identified as the pathologic hallmark of all three syndromes, and MSA was recognized as part of the pathologic family of neurodegenerative disorders termed "synucleinopathies" [7,8].

Beginning in the late 1990s, consensus groups developed clinical guidelines to define MSA, which were modified in 2008 and again in 2022 [6,9-12]. (See "Multiple system atrophy: Clinical features and diagnosis", section on 'Diagnostic criteria'.)

EPIDEMIOLOGY — MSA is a rare disease. The estimated annual incidence is 0.6 cases per 100,000 person-years [13]. In adults >50 years of age, the incidence increases to 3 per 100,000 person-years [14].

The estimated prevalence of MSA ranges from 2 to 5 cases per 100,000 population [13,15,16]. These estimates are approximately 10-fold lower compared with those of Parkinson disease (PD). (See "Epidemiology, pathogenesis, and genetics of Parkinson disease", section on 'Epidemiology'.)

The only established risk factor for MSA is advancing age. The median age of symptom onset is in the sixth decade, which is approximately five years younger than that of PD. (See "Multiple system atrophy: Clinical features and diagnosis", section on 'Age of onset'.)

Males and females are affected in approximately equal proportions [17-20], although some studies have reported a male predominance [21,22]. This finding may be secondary to earlier recognition of impotence as a major diagnostic feature in males.

There are no specific racial/ethnic predilections, and the disease has been reported worldwide. Rare families with MSA have been described, but the majority of cases appear to be sporadic. (See 'Genetics' below.)

There are no established environmental risk factors for MSA, although data are limited by its rarity [8,23,24].

PATHOLOGY

Hallmark features — MSA is a synucleinopathy resulting in neuronal loss and atrophy in basal ganglia, brainstem nuclei, cerebellum, and corticospinal tracts. Cytoplasmic aggregates of alpha-synuclein in oligodendrocytes, referred to as glial cytoplasmic inclusions, are the pathologic hallmark. The involvement of glial cells distinguishes MSA from other synucleinopathies such as Parkinson disease (PD), in which alpha-synuclein aggregates (Lewy bodies) are present in neurons. (See "Epidemiology, pathogenesis, and genetics of Parkinson disease", section on 'Pathology'.)

Alpha-synuclein immunostaining is a sensitive marker of inclusion pathology in MSA [8,25]. In addition to hyperphosphorylated alpha-synuclein, the glial cytoplasmic inclusions also contain tau, ubiquitin, leucine-rich repeat serine/threonine-protein kinase 2, and many other proteins [26-30]. Neuronal inclusions of various types, including Lewy body-like inclusions, are also present in the majority of patients [31,32].

The fibrillary structure of alpha-synuclein aggregates in MSA is distinct from that of other synucleinopathies, which may explain cell and region-specific vulnerabilities as well as differences in phenotype [33-36]. The secondary beta-sheet structure of folded alpha-synuclein filaments is distinct in MSA compared with fibrils found in Lewy bodies. Content of the beta-sheet structure in glial cytoplasmic inclusions is also lower than what is measured in Lewy bodies [37].

Neuropathologic distribution — The clinical presentation of MSA is largely determined by the pathologic distribution of glial cytoplasmic inclusions, along with the degree of neuronal loss within specific regions of the neuraxis. Neuronal cell loss correlates with the pathologic burden of glial cytoplasmic inclusions and disease duration [38]. Typical sites of pathologic involvement in MSA include the putamen, caudate nucleus, substantia nigra, locus ceruleus, pontine nuclei, inferior olivary nucleus, Purkinje cell layer of the cerebellum, and intermediolateral cell columns [39].

The pattern and degree of neuropathologic changes show some correlation with clinical subtype. Early changes in patients with predominant parkinsonism (MSA-P) are in the caudate, putamen, globus pallidus, and substantia nigra, whereas patients with predominant cerebellar ataxia (MSA-C) show more prominent cell loss in the cerebellum, pons, inferior olives, substantia nigra, and cerebral hemispheres [40,41]. (See "Multiple system atrophy: Clinical features and diagnosis", section on 'Motor involvement'.)

PATHOGENESIS — The cause of MSA is unknown [42]. However, the presence of alpha-synuclein messenger RNA (mRNA [ribonucleic acid]) in oligodendrocytes as well as glial inclusions and myelin disruption in the central nervous system suggest that MSA is a primary disorder of the glia [42-44].

Mechanisms — The mechanisms by which alpha-synuclein aggregates participate in the neurodegenerative cascade are under investigation.

One postulated mechanism involves prion-like spreading of aberrant alpha-synuclein from neurons to glia in MSA through functionally connected networks, thereby leading to glial and myelin dysfunction and an inflammatory cascade that promotes secondary neurodegeneration [45]. There have been a number of experimental studies demonstrating that abnormal alpha-synuclein aggregates might be responsible for the progression of MSA [46,47]. However, this remains controversial and is still an area of investigation [44,48].

Even before inclusions appear, oligodendrocytes undergo significant morphologic and biochemical changes, including cell enlargement, cytoplasmic clearing, and aggregation of tubulin polymerization-promoting phosphoprotein-25-alpha (p25α). p25a associates with myelin basic protein and moves from the nucleus and cell processes to the cytoplasm in oligodendrocytes prior to aggregation of alpha-synuclein [49,50].

Myelin degeneration is characteristic of MSA. A small case-control study found a significantly greater degree of white matter hyperintensities on magnetic resonance imaging (MRI) scans from patients with MSA compared with scans from patients with Parkinson disease (PD) and healthy controls [51]. This finding could be related to the dysfunction and loss of myelin or to cerebral hypoperfusion from orthostatic blood pressure fluctuations in MSA.

Neuroinflammation may also contribute to neurodegeneration in MSA. Increased numbers of activated microglia and astrocytes have been observed in autopsy specimens of patients with MSA, including microglia expressing nucleotide-binding domain, leucine-rich repeats-containing family, pyrin domain-containing-3 (NLRP3) inflammasome-related proteins in the putamen [52]. Further studies suggest that the inflammatory cascade may be propagated by interaction between alpha-synuclein and the NLRP3 inflammasome, leading to caspase 1 activation [53,54].

Clinical correlations — MSA by definition involves degeneration of multiple systems, which contribute to the varying clinical phenotypes of the disease. (See "Multiple system atrophy: Clinical features and diagnosis", section on 'Clinical features'.)

Autonomic dysfunction – Autonomic dysfunction in MSA is secondary to loss of cells in the intermediolateral cell columns [55,56] and loss of catecholaminergic neurons of the C1 area of the ventrolateral medulla (VLM) [57]. This is manifested by severe variability in blood pressure and heart rate, orthostatic hypotension, syncope, and postprandial hypotension. Arginine-vasopressin release from magnocellular hypothalamic neurons is impaired; this may be mediated by loss of A1 neurons in the caudal VLM.

The peripheral autonomic system appears to be spared in MSA [58]. Some persistence of autonomic tone may be responsible for the frequently observed supine hypertension in MSA. By contrast, dysautonomia in PD is caused by peripheral nervous system dysfunction, particularly myocardial sympathetic denervation [59].

Mechanisms leading to urinary dysfunction include detrusor hyperreflexia from loss of inhibitory input to the pontine micturition center, loss of corticotropin-releasing factor neurons in the pontine micturition area [57], and bladder atonia primarily from severe damage to Onuf's nucleus in the sacral spinal cord. Damage to Onuf's nucleus is also the most likely cause of erectile dysfunction in males.

Parkinsonism – Motor abnormalities seen in MSA with predominant parkinsonism (MSA-P) are due primarily to neuronal loss and gliosis in the substantia nigra, putamen, caudate, and globus pallidus [4]. One of the features that distinguishes MSA and other atypical parkinsonian syndromes (ie, Parkinson-plus syndromes) from idiopathic PD is the lack of dramatic and sustained response to levodopa, although in early MSA such a response may be seen temporarily. The extent of putaminal involvement may determine the poor response to levodopa [38].

Cerebellar dysfunction – The cerebellar ataxia and pyramidal signs that characterize the MSA are secondary to degeneration of the cerebellar Purkinje cells, middle cerebellar peduncles, inferior olivary nuclei, basis pontis, and pontine nuclei. However, a majority of patients with predominant cerebellar ataxia (MSA-C) probably have subclinical loss of nigral neurons based upon findings from 123I-FP-CIT single-photon emission computed tomography (SPECT) imaging [60].

Disordered sleep and breathing – Loss of cholinergic mesopontine neurons, combined with loss of locus ceruleus neurons and preservation of rostral raphe neurons, may contribute to rapid eye movement (REM) sleep abnormalities often seen in MSA [57].

Respiratory abnormalities may reflect loss of cholinergic neurons in the arcuate nucleus of the ventral medulla [57]. Respiratory stridor, abnormal nocturnal ventilation, and pseudobulbar features are possibly secondary to brainstem pathology, and may involve the nucleus ambiguus [8].

GENETICS — The role of genetics in the pathogenesis of MSA is undefined, and the vast majority of cases appear to be sporadic. One report found that variants in the coenzyme Q2, polyprenyltransferase (COQ2) gene were associated with familial MSA in two Japanese families [61]. Other reports have described familial patterns of MSA but have not identified a causative genetic variant [62-64].

Genetic variants in the leucine-rich repeat kinase 2 (LRRK2) gene and the glucocerebrosidase (GBA1) gene, both common genetic forms of Parkinson disease, can present as MSA clinically as well as neuropathologically [65-67]. Another study identified a coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) pathogenic variant in a patient with MSA [68]. (See "Epidemiology, pathogenesis, and genetics of Parkinson disease", section on 'Monogenic forms of PD'.)

Major risk alleles have not yet been identified, or studies have been conflicting. A genome-wide association study from Europe and North America that compared 918 patients with MSA and 3864 controls found no significant risk loci and, in particular, did not find an association of MSA with genetic variation in COQ2 or synuclein alpha (SNCA) [69], which had been reported in earlier studies [70-72]. Rather, the microtubule-associated protein tau (MAPT) was the only gene that appeared to be associated with MSA, and two other studies suggest that MAPT haplotypes H1x and H1 may be associated with an increased risk of MSA [73,74]. The role of tau isoforms in MSA is currently unknown.

SUMMARY

Nomenclature – Multiple system atrophy (MSA) is an adult-onset neurodegenerative disorder of the basal ganglia, brainstem, cerebellum, and spinal cord that manifests with various combinations of autonomic dysfunction, parkinsonism, ataxia, and corticospinal degeneration.

MSA is a unifying term that brings together syndromes that used to be referred to as olivopontocerebellar atrophy (OPCA), striatonigral degeneration, and Shy-Drager syndrome. (See 'History and nomenclature' above.)

Epidemiology – The estimated annual incidence of MSA in the population >50 years old is approximately 3 per 100,000 person-years. The mean age of onset ranges from 54 to 60 years. There appears to be no racial or sex predilection. (See 'Epidemiology' above.)

Pathology – Cytoplasmic aggregates of alpha-synuclein in oligodendrocytes, referred to as glial cytoplasmic inclusions, are the pathologic hallmark of MSA. The inclusions also contain tau and ubiquitin. (See 'Hallmark features' above.)

Typical sites of pathologic involvement include the putamen, caudate nucleus, substantia nigra, locus ceruleus, pontine nuclei, inferior olivary nucleus, Purkinje cell layer of the cerebellum, and intermediolateral cell columns. (See 'Neuropathologic distribution' above.)

Pathogenesis – The cause of MSA is unknown, but most evidence points to the disease being a primary disorder of the glia. One postulated mechanism involves prion-like spreading of aberrant alpha-synuclein through functionally connected networks. (See 'Pathogenesis' above.)

Genetics – The genetic basis of MSA is not well understood, and the vast majority of cases appear to be sporadic. (See 'Genetics' above.)

  1. Dejerine J, Thomas A. L'atrophie olivo-ponto-cérébelleuse. Nouvelle iconographie de la Salpêtrière 1900; 13:330.
  2. Bradbury S, Eggleston C. Postural hypotension: a report of three cases. American Heart Journal 1925; 1:73.
  3. SHY GM, DRAGER GA. A neurological syndrome associated with orthostatic hypotension: a clinical-pathologic study. Arch Neurol 1960; 2:511.
  4. van der Eecken H, Adams RD, van Bogaert L. Striopallidal-nigral degeneration. An hitherto undescribed lesion in paralysis agitans. J Neuropathol Exp Neurol 1960; 19:159.
  5. Graham JG, Oppenheimer DR. Orthostatic hypotension and nicotine sensitivity in a case of multiple system atrophy. J Neurol Neurosurg Psychiatry 1969; 32:28.
  6. Gilman S, Low PA, Quinn N, et al. Consensus statement on the diagnosis of multiple system atrophy. J Neurol Sci 1999; 163:94.
  7. Papp MI, Kahn JE, Lantos PL. Glial cytoplasmic inclusions in the CNS of patients with multiple system atrophy (striatonigral degeneration, olivopontocerebellar atrophy and Shy-Drager syndrome). J Neurol Sci 1989; 94:79.
  8. Wenning GK, Colosimo C, Geser F, Poewe W. Multiple system atrophy. Lancet Neurol 2004; 3:93.
  9. Consensus statement on the definition of orthostatic hypotension, pure autonomic failure, and multiple system atrophy. The Consensus Committee of the American Autonomic Society and the American Academy of Neurology. Neurology 1996; 46:1470.
  10. Gilman S, Low P, Quinn N, et al. Consensus statement on the diagnosis of multiple system atrophy. American Autonomic Society and American Academy of Neurology. Clin Auton Res 1998; 8:359.
  11. Gilman S, Wenning GK, Low PA, et al. Second consensus statement on the diagnosis of multiple system atrophy. Neurology 2008; 71:670.
  12. Wenning GK, Stankovic I, Vignatelli L, et al. The Movement Disorder Society Criteria for the Diagnosis of Multiple System Atrophy. Mov Disord 2022; 37:1131.
  13. Bjornsdottir A, Gudmundsson G, Blondal H, Olafsson E. Incidence and prevalence of multiple system atrophy: a nationwide study in Iceland. J Neurol Neurosurg Psychiatry 2013; 84:136.
  14. Bower JH, Maraganore DM, McDonnell SK, Rocca WA. Incidence of progressive supranuclear palsy and multiple system atrophy in Olmsted County, Minnesota, 1976 to 1990. Neurology 1997; 49:1284.
  15. Schrag A, Ben-Shlomo Y, Quinn NP. Prevalence of progressive supranuclear palsy and multiple system atrophy: a cross-sectional study. Lancet 1999; 354:1771.
  16. Tison F, Yekhlef F, Chrysostome V, Sourgen C. Prevalence of multiple system atrophy. Lancet 2000; 355:495.
  17. Geser F, Seppi K, Stampfer-Kountchev M, et al. The European Multiple System Atrophy-Study Group (EMSA-SG). J Neural Transm (Vienna) 2005; 112:1677.
  18. Wüllner U, Schmitz-Hübsch T, Abele M, et al. Features of probable multiple system atrophy patients identified among 4770 patients with parkinsonism enrolled in the multicentre registry of the German Competence Network on Parkinson's disease. J Neural Transm (Vienna) 2007; 114:1161.
  19. Köllensperger M, Geser F, Ndayisaba JP, 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.
  20. Wenning GK, Geser F, Krismer F, et al. The natural history of multiple system atrophy: a prospective European cohort study. Lancet Neurol 2013; 12:264.
  21. Wenning GK, Ben Shlomo Y, Magalhães M, et al. Clinical features and natural history of multiple system atrophy. An analysis of 100 cases. Brain 1994; 117 ( Pt 4):835.
  22. Vanacore N. Epidemiological evidence on multiple system atrophy. J Neural Transm (Vienna) 2005; 112:1605.
  23. Nee LE, Gomez MR, Dambrosia J, et al. Environmental-occupational risk factors and familial associations in multiple system atrophy: a preliminary investigation. Clin Auton Res 1991; 1:9.
  24. Vidal JS, Vidailhet M, Elbaz A, et al. Risk factors of multiple system atrophy: a case-control study in French patients. Mov Disord 2008; 23:797.
  25. Wakabayashi K, Kawachi I, Toyoshima Y, Takahashi H. [Occurrence of argyrophilic grains in multiple system atrophy: histopathological examination of 26 autopsy cases]. No To Shinkei 1999; 51:433.
  26. Cairns NJ, Atkinson PF, Hanger DP, et al. Tau protein in the glial cytoplasmic inclusions of multiple system atrophy can be distinguished from abnormal tau in Alzheimer's disease. Neurosci Lett 1997; 230:49.
  27. Dickson DW, Liu W, Hardy J, et al. Widespread alterations of alpha-synuclein in multiple system atrophy. Am J Pathol 1999; 155:1241.
  28. Irving EA, Nicoll J, Graham DI, Dewar D. Increased tau immunoreactivity in oligodendrocytes following human stroke and head injury. Neurosci Lett 1996; 213:189.
  29. Probst-Cousin S, Bergmann M, Kuchelmeister K, et al. Ubiquitin-positive inclusions in different types of multiple system atrophy: distribution and specificity. Pathol Res Pract 1996; 192:453.
  30. Jellinger KA, Lantos PL. Papp-Lantos inclusions and the pathogenesis of multiple system atrophy: an update. Acta Neuropathol 2010; 119:657.
  31. Cykowski MD, Coon EA, Powell SZ, et al. Expanding the spectrum of neuronal pathology in multiple system atrophy. Brain 2015; 138:2293.
  32. Papp MI, Lantos PL. The distribution of oligodendroglial inclusions in multiple system atrophy and its relevance to clinical symptomatology. Brain 1994; 117 ( Pt 2):235.
  33. Schweighauser M, Shi Y, Tarutani A, et al. Structures of α-synuclein filaments from multiple system atrophy. Nature 2020; 585:464.
  34. Shahnawaz M, Mukherjee A, Pritzkow S, et al. Discriminating α-synuclein strains in Parkinson's disease and multiple system atrophy. Nature 2020; 578:273.
  35. Yang Y, Shi Y, Schweighauser M, et al. Structures of α-synuclein filaments from human brains with Lewy pathology. Nature 2022; 610:791.
  36. Van der Perren A, Gelders G, Fenyi A, et al. The structural differences between patient-derived α-synuclein strains dictate characteristics of Parkinson's disease, multiple system atrophy and dementia with Lewy bodies. Acta Neuropathol 2020; 139:977.
  37. Araki K, Yagi N, Ikemoto Y, et al. The secondary structural difference between Lewy body and glial cytoplasmic inclusion in autopsy brain with synchrotron FTIR micro-spectroscopy. Sci Rep 2020; 10:19423.
  38. Ozawa T, Paviour D, Quinn NP, et al. The spectrum of pathological involvement of the striatonigral and olivopontocerebellar systems in multiple system atrophy: clinicopathological correlations. Brain 2004; 127:2657.
  39. Lantos PL. The definition of multiple system atrophy: a review of recent developments. J Neuropathol Exp Neurol 1998; 57:1099.
  40. 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.
  41. Campese N, Fanciulli A, Stefanova N, et al. Neuropathology of multiple system atrophy: Kurt Jellinger`s legacy. J Neural Transm (Vienna) 2021; 128:1481.
  42. Wenning GK, Stefanova N, Jellinger KA, et al. Multiple system atrophy: a primary oligodendrogliopathy. Ann Neurol 2008; 64:239.
  43. Matsuo A, Akiguchi I, Lee GC, et al. Myelin degeneration in multiple system atrophy detected by unique antibodies. Am J Pathol 1998; 153:735.
  44. Meissner WG, Fernagut PO, Dehay B, et al. Multiple System Atrophy: Recent Developments and Future Perspectives. Mov Disord 2019; 34:1629.
  45. Jellinger KA. Neuropathology of multiple system atrophy: new thoughts about pathogenesis. Mov Disord 2014; 29:1720.
  46. Prusiner SB, Woerman AL, Mordes DA, et al. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc Natl Acad Sci U S A 2015; 112:E5308.
  47. Woerman AL, Kazmi SA, Patel S, et al. MSA prions exhibit remarkable stability and resistance to inactivation. Acta Neuropathol 2018; 135:49.
  48. Heras-Garvin A, Wenning GK. Is Multiple System Atrophy a New Prion Disorder? Mov Disord 2016; 31:300.
  49. Song YJ, Lundvig DM, Huang Y, et al. p25alpha relocalizes in oligodendroglia from myelin to cytoplasmic inclusions in multiple system atrophy. Am J Pathol 2007; 171:1291.
  50. Ota K, Obayashi M, Ozaki K, et al. Relocation of p25α/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. Umoto M, Miwa H, Ando R, et al. White matter hyperintensities in patients with multiple system atrophy. Parkinsonism Relat Disord 2012; 18:17.
  52. Li F, Ayaki T, Maki T, et al. NLRP3 Inflammasome-Related Proteins Are Upregulated in the Putamen of Patients With Multiple System Atrophy. J Neuropathol Exp Neurol 2018; 77:1055.
  53. Pike AF, Varanita T, Herrebout MAC, et al. α-Synuclein evokes NLRP3 inflammasome-mediated IL-1β secretion from primary human microglia. Glia 2021; 69:1413.
  54. Trudler D, Nazor KL, Eisele YS, et al. Soluble α-synuclein-antibody complexes activate the NLRP3 inflammasome in hiPSC-derived microglia. Proc Natl Acad Sci U S A 2021; 118.
  55. Gray F, Vincent D, Hauw JJ. Quantitative study of lateral horn cells in 15 cases of multiple system atrophy. Acta Neuropathol 1988; 75:513.
  56. Oppenheimer DR. Lateral horn cells in progressive autonomic failure. J Neurol Sci 1980; 46:393.
  57. Benarroch EE. Brainstem in multiple system atrophy: clinicopathological correlations. Cell Mol Neurobiol 2003; 23:519.
  58. Parikh SM, Diedrich A, Biaggioni I, Robertson D. The nature of the autonomic dysfunction in multiple system atrophy. J Neurol Sci 2002; 200:1.
  59. Goldstein DS, Holmes CS, Dendi R, et al. Orthostatic hypotension from sympathetic denervation in Parkinson's disease. Neurology 2002; 58:1247.
  60. Muñoz E, Iranzo A, Rauek S, et al. Subclinical nigrostriatal dopaminergic denervation in the cerebellar subtype of multiple system atrophy (MSA-C). J Neurol 2011; 258:2248.
  61. Multiple-System Atrophy Research Collaboration. Mutations in COQ2 in familial and sporadic multiple-system atrophy. N Engl J Med 2013; 369:233.
  62. Hara K, Momose Y, Tokiguchi S, et al. Multiplex families with multiple system atrophy. Arch Neurol 2007; 64:545.
  63. Wüllner U, Schmitt I, Kammal M, et al. Definite multiple system atrophy in a German family. J Neurol Neurosurg Psychiatry 2009; 80:449.
  64. Hohler AD, Singh VJ. Probable hereditary multiple system atrophy-autonomic (MSA-A) in a family in the United States. J Clin Neurosci 2012; 19:479.
  65. Lee K, Nguyen KD, Sun C, et al. LRRK2 p.Ile1371Val Mutation in a Case with Neuropathologically Confirmed Multi-System Atrophy. J Parkinsons Dis 2018; 8:93.
  66. Riboldi GM, Palma JA, Cortes E, et al. Early-onset pathologically proven multiple system atrophy with LRRK2 G2019S mutation. Mov Disord 2019; 34:1080.
  67. Sklerov M, Kang UJ, Liong C, et al. Frequency of GBA variants in autopsy-proven multiple system atrophy. Mov Disord Clin Pract 2017; 4:574.
  68. Nicoletti G, Gagliardi M, Procopio R, et al. A new CHCHD2 mutation identified in a southern italy patient with multiple system atrophy. Parkinsonism Relat Disord 2018; 47:91.
  69. Sailer A, Scholz SW, Nalls MA, et al. A genome-wide association study in multiple system atrophy. Neurology 2016; 87:1591.
  70. Sasaki H, Emi M, Iijima H, et al. Copy number loss of (src homology 2 domain containing)-transforming protein 2 (SHC2) gene: discordant loss in monozygotic twins and frequent loss in patients with multiple system atrophy. Mol Brain 2011; 4:24.
  71. Scholz SW, Houlden H, Schulte C, et al. SNCA variants are associated with increased risk for multiple system atrophy. Ann Neurol 2009; 65:610.
  72. Al-Chalabi A, Dürr A, Wood NW, et al. Genetic variants of the alpha-synuclein gene SNCA are associated with multiple system atrophy. PLoS One 2009; 4:e7114.
  73. Labbé C, Heckman MG, Lorenzo-Betancor O, et al. MAPT haplotype diversity in multiple system atrophy. Parkinsonism Relat Disord 2016; 30:40.
  74. Vilariño-Güell C, Soto-Ortolaza AI, Rajput A, et al. MAPT H1 haplotype is a risk factor for essential tremor and multiple system atrophy. Neurology 2011; 76:670.
Topic 139535 Version 1.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟