Skip Navigation
Skip to contents

JMD : Journal of Movement Disorders

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Mov Disord > Volume 3(1); 2010 > Article
Review Article
Clinicopathological Correlates of Lewy Body Disease: Fundamental Issues
Tae-Beom Ahn
Journal of Movement Disorders 2010;3(1):11-14.
DOI: https://doi.org/10.14802/jmd.10003
Published online: April 30, 2010

Department of Neurology, School of Medicine, Kyung Hee University, Seoul, Korea

Corresponding author: Tae-Beom Ahn, MD, PhD, Department of Neurology, School of Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-702, Korea, Tel +82-2-958-8448, Fax +82-2-958-8495, E-mail ricash@hanmail.net
• Received: April 10, 2010   • Accepted: April 20, 2010

Copyright © 2010 The Korean Movement Disorder Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 10,398 Views
  • 62 Download
  • 3 Crossref
  • Lewy body pathology (LBP) is the pathological hallmark of Lewy body diseases, such as Parkinson’s disease and Lewy body dementia. Recent studies have shed new light on the role of LBP, the interactions of LBP with concomitant pathologies, and the propagation of LBP from the olfactory bulb and enteric nervous system to the central nervous system. The intrinsic difficulty with identifying clinicopathological correlates could be overcome by improving our understanding of the pathological evolution of LBP.
Lewy body pathology (LBP) characterizes neurodegenerative diseases such as Parkinson’s disease (PD) and Lewy body dementia (DLB). Since the seminal report on PD by James Parkinson, Lewy bodies (LBs), especially in the substantia nigra (SN), have drawn the attention of many researchers, and the presence of LBs in the SN has been considered highly specific for the pathological diagnosis of PD.1,2 Japanese researchers introduced the idea of diffuse Lewy body disease (LBD) to describe the remarkable distribution of LBP across the cortices.35 Later studies followed their suggestions and refined the diagnostic procedure, establishing clinical and pathological criteria for DLB.6
On hematoxylin-eosin staining, many LBs in the brainstem consist of an eosinophilic core and peripheral halo, whereas they usually appear as irregular eosinophilic inclusions in other limbic and cortical areas.7 Of the numerous proteins contained in LBs, α-synuclein (aSyn) is the most abundant, and many of the proteins are ubiquitinated.7 The central predominance of aSyn and the peripheral halo of densely ubiquitinated proteins is a conspicuous feature of brainstem LB.
The organized feature of brainstem LBs implies order in the process of LB formation. The aggresome hypothesis holds that the failure of the aggresome to remove unwanted proteins is a primordial event.8 A LB might form from a failed aggresome, but not in a haphazard way. Consequently, LB formation could be an ‘active’ process designed to segregate harmful proteins from the neurons.
The pathogenesis of PD has been discussed in association with cellular machinery such as the proteasome and autophagy for the removal of unwanted proteins.9,10 Direct evidence of this came from human brain, which showed structural defects in, and functional impairment of, the proteasome.1113 Experimental studies recapitulated neuronal death and the formation of aSyn aggregates by proteasomal inhibition, which could be considered as a forme fruste of LB.14,15 Autophagy is another important system for dealing with toxic waste. Interestingly, chaperone-mediated autophagy was hampered by a mutant aSyn or aSyn-dopa adducts, forming aSyn aggregates.16 These data suggest that the formation of the LB is closely related to the impairment of major intracellular machinery. However, the ultimate upstream mechanism responsible for the active regulation of the machinery that handles toxic waste by segregating it into aggregates is still under investigation.
The neuroprotection conferred by LB formation has been studied in the human brain using in-situ end-labeling for apoptosis, in which the neurons with LBs were less frequently apoptotic than those without.17 Another study showed the downregulation of tyrosine hydroxylase (TH) in the neurons harboring LBs.18 The presence of LBs might alert the neurons to prepare for the threat to come. As TH is a key enzyme in the metabolism of dopamine in the SN, and dopamine oxidation can produce highly toxic products, such as dopamine adducts, TH downregulation could be an effective compensatory way to survive.
As LBP is commonly found in a background of severe neuronal loss, as in the case of PD, LBP cannot be easily exempted from neuron death. A recent pathological study showed a stable proportion of LB-containing neurons in the SN, suggesting a balance between the formation and removal of LBs.19 The authors calculated the life span of neurons with LB to be 6.2 months versus 15.9 months for those with any type of aSyn inclusion. The limited life span of the neurons carrying LBs argues for the detrimental role of LBs.
Without prospective data, it cannot be determined whether LB formation signifies a tombstone for a cell death cascade or a successful salvage operation to sequester toxicants. However, recent studies have partly suggested that the LB is not necessarily a culprit in neuronal death.
Incidental Lewy body disease (iLBD) is a pathological entity defined by the presence of aSyn pathology without any clinical evidence of PD or DLB. As iLBD is found in normal persons, by definition, its pathology offers invaluable insight into the early evolution of LBD, such as PD and DLB.2022 Moreover, although iLBD has no clinical implications, by definition, it is significantly associated with a clinical prodrome of PD that includes olfactory dysfunction and bowel frequency, which provide a pathological basis for the early diagnosis of PD.23,24
In iLBD, the neuronal loss is usually minimal, despite aSyn pathology.2022 The dissociation between neuronal loss and aSyn pathology contrasts the marked neuronal death and widespread LBP in the SN in PD, which suggests that LBP is less toxic than expected, at least in iLBD. Then, if iLBD is a true prodrome of PD and its pathology recapitulates the early evolution of the LBP of PD, the transition from iLBD to PD may be made by additional provoking factors, rather than by a simple shift in the continuum of LBD.
When microgliosis and astrogliosis are studied in the brainstem nuclei, including the dorsal motor nucleus of the vagus nerve (dmV) in the medulla, locus ceruleus (LC) in the pons, and SN in the midbrain, neuroinflammation is only increased slightly in iLBD, compared to normal controls, whereas inflammatory change is conspicuous in PD.25 Interestingly, although the inflammatory change was similar in the three brainstem nuclei in iLBD, the SN was more severely affected by neuroinflammation than the dmV and LC in PD. The drastic change in inflammation might underlie the pattern shift of LBP from iLBD to PD. Other potential factors contributing to the pattern shift are a high iron content, excessive oxidative stress, and impaired anti-oxidative mechanism.2628 The disproportionate involvement of the SN is in line with the conspicuous motor manifestations of PD, closely associated with SN pathology, while no clinical symptoms are present in iLBD.
The recent hypothesis on the caudo-rostral propagation of LBP suggested by Braak assumes that the appearance of aSyn pathology in SN is a key feature of entering the symptomatic phase.29,30 However, there are many iLBD cases with an extranigral distribution, even involving the neocortices.22 In fact, some cases with iLBD could be classified as a DLB-like group, due to the widespread distribution of aSyn pathology. iLBD could be ‘pro-PD’ (brainstem predominant iLBD) or ‘pro-DLB’ (diffuse iLBD).22 These findings suggest the occurrence of LBP across the brain without a significant temporal gap, not respecting the border between the brainstem and cortices, rather than in a consecutive manner from the lower brainstem to the higher cortices, without skipping intervening tissues.
Another difficult problem is that iLBD is not exclusively related to LBD. Other neurodegenerative diseases can accompany LBP, such as Alzheimer’s disease (AD) and progressive supranuclear palsy.31,32 A small proportion of the patients with multiple system atrophy and corticobasal degeneration also have associated LBD.33 As LBP is found in normal subjects and patients with various neurodegenerative diseases, it is possible to say that LBD might develop in any brain, regardless of the concomitant pathologies.31 This again raises a vexing question about the functional contribution of LBP.
To understand the functional role of LBP, it is necessary to have at least two sets of data: one concerning the quantitative relationship between the clinical features and the burden of LBP, and the other concerning the interaction with other concomitant pathologies, to isolate the ‘pure’ contribution of LBP. Surprisingly, although cognitive function was reported to be correlated with cortical LBs, few studies have succeeded in showing a significant relationship between Parkinsonism and aSyn pathology. A recent prospective study showed that Parkinsonism was significantly correlated with the burden of aSyn pathology. 34
Facing the problem of mixed AD and LB pathology in DLB, researchers adopted the concept of probability to discriminate the functional significance of the specific pathologies.6 The cases heavily loaded with AD pathology are less likely to be diagnosed as DLB, even with diffuse LBD according to the published consensus criteria. Recently, Obi et al.35 studied β-amyloid, tau protein, and LBP together and showed the central role of β-amyloid in the evolution of tau pathology. LBP was not entirely dependent on β-amyloid pathology.35 These attempts underscore the importance of sifting through concomitant pathologies to identify the independent contribution of LBP.
Apart from AD pathologies such as tau [neurofibrillary tangle (NFT)] and β-amyloid [senile plaque (SP)], other pathologies might modify the clinicopathological presentation of LBP; these include, vascular pathologies, argyrophilic grain disease (AGD) and TAR DNA binding protein-43 (TDP43).3639 AGD and TDP43 could contribute to cognitive decline, which would be difficult to discern in the context of coexisting limbic AD pathologies or cortical LBs. Vascular pathologies have a more complicated implication because they show great variability in terms of the amount and distribution of the lesions. A comprehensive approach that considers all of the coexisting pathologies together is needed to determine a full picture of the clinicopathological interactions, and to delineate the role of LBP.
Recent studies of fetal grafts demonstrated the propagation of the aSyn pathology from the host to the graft, which unraveled the contagious nature of aSyn pathology.40,41 Until recently, only grafts more than 10 years after transplantation were thought to be insinuated by aSyn pathologies, and earlier studies showed no aSyn pathologies in grafts younger than 10 years.4245
Several mechanisms were suggested to explain the human pathology.46 In a recent case, a graft was studied 14 years after transplantation.47 Although the graft was expected to be localized in the putamen, it had extended from the putamen to the amygdala. aSyn inclusions were found in the graft, and they were more frequent in the portion lying in the amygdala. Astrocytosis and microgliosis were also more prominent in the amygdala portion of the graft. The myelination of the graft was poor and mainly found in the putaminal portion. Corpora amylacea (as a marker of aging) was more common in the peripheral portion of the graft. These findings indicated that the maturation and aging of the graft might not be critical to the development of aSyn pathology. As the amygdala is more severely affected than the putamen in LBD, and could frequently be affected by various abnormal protein aggregations, such as NFTs, TDP-43, and argyrophilic grains, the amygdala might have a greater tendency to develop pathological inclusions than the putamen. 36,48,49 In other words, the putamen-amygdala gradient of aSyn inclusions might be affected by factors constituting the ‘aggregation-prone’ milieu of the amygdala, which remains speculative. Recently, cell-to-cell propagation of aSyn inclusions was demonstrated experimentally.50 As the pathologic burden of aSyn was heavier in the amygdala, the propagation might explain the putamen-amygdala gradient. Direct propagation from the neighboring tissue might produce a center-peripheral gradient of aSyn inclusions in the graft, which was, however, not observed in our case.
After its birth in iLBD, LBP might ‘mature’, affecting various regions with or without concomitant pathologies. It remains controversial whether LBP spreads from limited pathological foci (the enteric nervous system or olfactory bulb according to the hypothesis of Braak) or arises nearly simultaneously, but disproportionately, in multiple sites.22,30 In some cases of AD, only the amygdala is affected by LBP, sparing other brain areas involved in the classical forms of LBD, which argues for the latter scenario.51 Recent data showed that the explanatory power of Braak staging for LBP is variable, and is most compelling in LBD associated with progressive supranuclear palsy. 32 As LBP commonly accompanies other pathologies, its functional role should be dissected from the influence of concomitant pathologies.

The author has no financial conflicts of interest.

  • 1. Forno LS. Neuropathology of Parkinson’s disease. J Neuropathol Exp Neurol 1996;55:259–272.ArticlePubMed
  • 2. Parkinson J. An essay on the shaking palsy. 1817. J Neuropsychiatry Clin Neurosci 2002;14:223–236.discussion 222.. ArticlePubMed
  • 3. Kosaka K. Lewy bodies in cerebral cortex, report of three cases. Acta Neuropathol 1978;42:127–134.ArticlePubMed
  • 4. Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy body disease: progressive dementia with abundant cortical Lewy bodies and senile changes of varying degree--a new disease? Clin Neuropathol 1984;3:185–192.PubMed
  • 5. Okazaki H, Lipkin LE, Aronson SM. Diffuse intracytoplasmic ganglionic inclusions (Lewy type) associated with progressive dementia and quadriparesis in flexion. J Neuropathol Exp Neurol 1961;20:237–244.ArticlePubMed
  • 6. McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al. Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium. Neurology 2005;65:1863–1872.ArticlePubMed
  • 7. Wakabayashi K, Tanji K, Mori F, Takahashi H. The Lewy body in Parkinson’s disease: molecules implicated in the formation and degradation of alpha-synuclein aggregates. Neuropathology 2007;27:494–506.ArticlePubMed
  • 8. Olanow CW, Perl DP, DeMartino GN, McNaught KS. Lewy-body formation is an aggresome-related process: a hypothesis. Lancet Neurol 2004;3:496–503.ArticlePubMed
  • 9. Cook C, Petrucelli L. A critical evaluation of the ubiquitin-proteasome system in Parkinson’s disease. Biochim Biophys Acta 2009;1792:664–675.ArticlePubMedPMC
  • 10. Yang Q, Mao Z. Parkinson Disease: a role for autophagy? Neuroscientist 2010.Article
  • 11. McNaught KS, Jenner P. Proteasomal function is impaired in substantia nigra in Parkinson’s disease. Neurosci Lett 2001;297:191–194.ArticlePubMed
  • 12. McNaught KS, Belizaire R, Jenner P, Olanow CW, Isacson O. Selective loss of 20S proteasome alpha-subunits in the substantia nigra pars compacta in Parkinson’s disease. Neurosci Lett 2002;326:155–158.ArticlePubMed
  • 13. McNaught KS, Belizaire R, Isacson O, Jenner P, Olanow CW. Altered proteasomal function in sporadic Parkinson’s disease. Exp Neurol 2003;179:38–46.ArticlePubMed
  • 14. McNaught KS, Mytilineou C, Jnobaptiste R, Yabut J, Shashidharan P, Jennert P, et al. Impairment of the ubiquitin-proteasome system causes dopaminergic cell death and inclusion body formation in ventral mesencephalic cultures. J Neurochem 2002;81:301–306.ArticlePubMed
  • 15. McNaught KS, Björklund LM, Belizaire R, Isacson O, Jenner P, Olanow CW. Proteasome inhibition causes nigral degeneration with inclusion bodies in rats. Neuroreport 2002;13:1437–1441.ArticlePubMed
  • 16. Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D. Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 2004;305:1292–1295.ArticlePubMed
  • 17. Tompkins MM, Hill WD. Contribution of somal Lewy bodies to neuronal death. Brain Res 1997;775:24–29.ArticlePubMed
  • 18. Mori F, Nishie M, Kakita A, Yoshimoto M, Takahashi H, Wakabayashi K. Relationship among alpha-synuclein accumulation, dopamine synthesis, and neurodegeneration in Parkinson disease substantia nigra. J Neuropathol Exp Neurol 2006;65:808–815.ArticlePubMed
  • 19. Greffard S, Verny M, Bonnet AM, Seilhean D, Hauw JJ, Duyckaerts C. A stable proportion of Lewy body bearing neurons in the substantia nigra suggests a model in which the Lewy body causes neuronal death. Neurobiol Aging 2010;31:99–103.ArticlePubMed
  • 20. DelleDonne A, Klos KJ, Fujishiro H, Ahmed Z, Parisi JE, Josephs KA, et al. Incidental Lewy body disease and preclinical Parkinson disease. Arch Neurol 2008;65:1074–1080.ArticlePubMed
  • 21. Dickson DW, Fujishiro H, DelleDonne A, Menke J, Ahmed Z, Klos KJ, et al. Evidence that incidental Lewy body disease is pre-symptomatic Parkinson’s disease. Acta Neuropathol 2008;115:437–444.ArticlePubMed
  • 22. Frigerio R, Fujishiro H, Ahn TB, Josephs KA, Maraganore DM, Delledonne A, et al. Incidental Lewy body disease: do some cases represent a preclinical stage of dementia with Lewy bodies? Neurobiol Aging 2009.ArticlePubMedPMC
  • 23. Ross GW, Abbott RD, Petrovitch H, Tanner CM, Davis DG, Nelson J, et al. Association of olfactory dysfunction with incidental Lewy bodies. Mov Disord 2006;21:2062–2067.ArticlePubMed
  • 24. Abbott RD, Ross GW, Petrovitch H, Tanner CM, Davis DG, Masaki KH, et al. Bowel movement frequency in late-life and incidental Lewy bodies. Mov Disord 2007;22:1581–1586.ArticlePubMed
  • 25. Ahn TB, Fujishiro H, Menke J, Frigerio R, DelleDonne A, Klos KJ, et al. Glial activation in incidental Lewy body disease. Parkinsonism Relat Disord 2009;15(supplement 2):S163.Article
  • 26. Litvan I, Halliday G, Hallett M, Goetz CG, Rocca W, Duyckaerts C, et al. The etiopathogenesis of Parkinson disease and suggestions for future research. Part I. J Neuropathol Exp Neurol 2007;66:251–257.ArticlePubMed
  • 27. Friedman A, Galazka-Friedman J, Koziorowski D. Iron as a cause of Parkinson disease-a myth or a well established hypothesis? Parkinsonism Relat Disord 2009;15( Suppl 3):S212–S214.ArticlePubMed
  • 28. Wypijewska A, Galazka-Friedman J, Bauminger ER, Wszolek ZK, Schweitzer KJ, Dickson DW, et al. Iron and reactive oxygen species activity in parkinsonian substantia nigra. Parkinsonism Relat Disord 2010.Article
  • 29. Braak H, Ghebremedhin E, Rüb U, Bratzke H, Del Tredici K. Stages in the development of Parkinson’s disease-related pathology. Cell Tissue Res 2004;318:121–134.ArticlePubMed
  • 30. Hawkes CH, Del Tredici K, Braak H. Parkinson’s disease: a dual-hit hypothesis. Neuropathol Appl Neurobiol 2007;33:599–614.ArticlePubMedPMC
  • 31. Uchikado H, DelleDonne A, Ahmed Z, Dickson DW. Lewy bodies in progressive supranuclear palsy represent an independent disease process. J Neuropathol Exp Neurol 2006;65:387–395.ArticlePubMed
  • 32. Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y. Evidence in favor of Braak staging of Parkinson’s disease. Mov Disord 2010;25( Suppl 1):S78–S82.ArticlePubMed
  • 33. Fujishiro H, Ahn TB, Frigerio R, Uchikado H, Klos KJ, Josephs KA, et al. Incidental Lewy bodies in various neurodegenerative disorders. Mov Disord 2008;23(S1):S30.Article
  • 34. Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, et al. Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol 2009;117:613–634.ArticlePubMedPMC
  • 35. Obi K, Akiyama H, Kondo H, Shimomura Y, Hasegawa M, Iwatsubo T, et al. Relationship of phosphorylated alpha-synuclein and tau accumulation to Abeta deposition in the cerebral cortex of dementia with Lewy bodies. Exp Neurol 2008;210:409–420.ArticlePubMed
  • 36. Ferrer I, Santpere G, van Leeuwen FW. Argyrophilic grain disease. Brain 2008;131:1416–1432.ArticlePubMed
  • 37. Higashi S, Iseki E, Yamamoto R, Minegishi M, Hino H, Fujisawa K, et al. Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res 2007;1184:284–294.ArticlePubMed
  • 38. Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy body disease. Acta Neuropathol 2008;115:427–436.ArticlePubMed
  • 39. Nakashima-Yasuda H, Uryu K, Robinson J, Xie SX, Hurtig H, Duda JE, et al. Co-morbidity of TDP-43 proteinopathy in Lewy body related diseases. Acta Neuropathol 2007;114:221–229.ArticlePubMed
  • 40. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson’s disease. Nat Med 2008;14:504–506.ArticlePubMed
  • 41. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. Lewy bodies in grafted neurons in subjects with Parkinson’s disease suggest host-to-graft disease propagation. Nat Med 2008;14:501–503.ArticlePubMed
  • 42. Kirik D, Björklund A. Histological analysis of fetal dopamine cell suspension grafts in two patients with Parkinson’s disease gives promising results. Brain 2005;128:1478–1479.ArticlePubMed
  • 43. Kordower JH, Freeman TB, Snow BJ, Vingerhoets FJ, Mufson EJ, Sanberg PR, et al. Neuropathological evidence of graft survival and striatal reinnervation after the transplantation of fetal mesencephalic tissue in a patient with Parkinson’s disease. N Engl J Med 1995;332:1118–1124.ArticlePubMed
  • 44. Kordower JH, Freeman TB, Chen EY, Mufson EJ, Sanberg PR, Hauser RA, et al. Fetal nigral grafts survive and mediate clinical benefit in a patient with Parkinson’s disease. Mov Disord 1998;13:383–393.ArticlePubMed
  • 45. Lee CC, Lin SZ, Wang Y, Lin JJ, Liu JY, Chen GJ, et al. First human ventral mesencephalon and striatum cografting in a Parkinson patient. Acta Neurochir Suppl 2003;87:159–162.ArticlePubMed
  • 46. Braak H, Del Tredici K. Assessing fetal nerve cell grafts in Parkinson’s disease. Nat Med 2008;14:483–485.ArticlePubMed
  • 47. Ahn TB, Fujishiro H, Menke J, Frigerio R, DelleDonne A, Klos KJ, et al. Relationship of neighboring tissue and gliosis to a-synuclein pathology in a fetal graft to treat Parkinson disease. Parkinsonism Relat Disord 2009;15(Supplement 2):S140.Article
  • 48. Chen-Plotkin AS, Lee VM, Trojanowski JQ. TAR DNA-binding protein 43 in neurodegenerative disease. Nat Rev Neurol 2010;6:211–220.ArticlePubMedPMC
  • 49. Duyckaerts C, Delatour B, Potiaer MC. Classification and basic pathology of Alzheimer disease. Acta Neuropathol 2009;118:5–36.ArticlePubMed
  • 50. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci U S A 2009;106:13010–13015.ArticlePubMedPMC
  • 51. Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol 2006;65:685–697.ArticlePubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Demencias degenerativas: ¿un dilema de síndromes o de enfermedades?
      A. Robles Bayón
      Neurología.2022; 37(6): 480.     CrossRef
    • Degenerative dementias: a question of syndrome or disease?
      A. Robles Bayón
      Neurología (English Edition).2022; 37(6): 480.     CrossRef
    • Advances in NURR1-Regulated Neuroinflammation Associated with Parkinson’s Disease
      Murad Al-Nusaif, Yushan Lin, Tianbai Li, Cheng Cheng, Weidong Le
      International Journal of Molecular Sciences.2022; 23(24): 16184.     CrossRef

    Comments on this article

    Add a comment

    JMD : Journal of Movement Disorders