ArticlePDF Available

First Appraisal of Brain Pathology Owing to A30P Mutant Alpha-Synuclein

Authors:

Abstract and Figures

Familial Parkinson disease (PD) due to the A30P mutation in the SNCA gene encoding alpha-synuclein is clinically associated with PD symptoms. In this first pathoanatomical study of the brain of an A30P mutation carrier, we observed neuronal loss in the substantia nigra, locus coeruleus, and dorsal motor vagal nucleus, as well as widespread occurrence of alpha-synuclein immunopositive Lewy bodies, Lewy neurites, and glial aggregates. Alpha-synuclein aggregates ultrastructurally resembled Lewy bodies, and biochemical analyses disclosed a significant load of insoluble alpha-synuclein, indicating neuropathological similarities between A30P disease patients and idiopathic PD, with a more severe neuropathology in A30P carriers.
Alpha-synuclein immunocytochemical and electron microscopic findings in the A30P index patient. (A) Lewy bodies (LBs) (large arrows) and Lewy neurites (arrowheads) in the dorsal motor vagal nucleus. Small arrows point to coiled bodies. (B) Gleason score glial fibrillary acidic protein (GFAP)-immunopositive astrocytes in the transentorhinal cortex containing alpha-synuclein immunopositive deposits (arrowheads). (C) Substantia nigra: Typical coiled-up alpha-synuclein immunopositive oligodendroglial inclusion (brown) surrounding the nucleus (blue) of the affected oligodendrocyte. (D) The dorsal motor vagal nucleus of the A30P patient exhibits various LBs (arrows) with a dark inner core and a ring-like light outer zone. (E, F) Electron micrograph of the LB in the lower left corner of D (asterisk). The LB shows a dense granular core (Co), surrounded by a lighter outer zone (OZ), which contains loosely arranged filaments (F). (G, H) Electron micrograph of an LB found in an idiopathic Parkinson disease case. Note the high structural similarity of the LBs from both cases. (A) Anti–alpha-synuclein immunocytochemistry; (B) anti-GFAP (3,3′-diaminobenzidine [DAB], brown)/anti–alpha-synuclein (SK4700, blue-gray) double immunostaining; (C) antitransferrin (SK4700, blue-gray)/anti–alpha-synuclein (DAB, brown) double immunostaining; (A–C) 100μm percutaneous endoscopic gastrostomy sections; (D) toluidine blue-stained semithin section, 0.75μm; (E–H) uranyl acetate and lead citrate contrasted ultrathin section, 100nm).
… 
Content may be subject to copyright.
First Appraisal of Brain
Pathology Owing to A30P
Mutant Alpha-Synuclein
Kay Seidel, PhD,
1
Ludger Scho¨ ls, MD,
2
Silke Nuber, PhD,
3
Elisabeth Petrasch-Parwez, MD,
4
Kristin Gierga, MD,
5
Zbigniew Wszolek, MD,
6
Dennis Dickson, MD,
7
Wei P. Gai, PhD, MD,
8
Antje Bornemann, MD,
9
Olaf Riess, MD,
3
Abdelhaq Rami, PhD,
10
Wilfried F. A. den Dunnen, MD,
11
Thomas Deller, MD,
1
Udo Ru¨b, MD,
1
and
Rejko Kru¨ ger, MD
2
Familial Parkinson disease (PD) due to the A30P
mutation in the SNCA gene encoding alpha-
synuclein is clinically associated with PD symp-
toms. In this first pathoanatomical study of the
brain of an A30P mutation carrier, we observed
neuronal loss in the substantia nigra, locus coer-
uleus, and dorsal motor vagal nucleus, as well as
widespread occurrence of alpha-synuclein immu-
nopositive Lewy bodies, Lewy neurites, and glial
aggregates. Alpha-synuclein aggregates ultra-
structurally resembled Lewy bodies, and biochem-
ical analyses disclosed a significant load of insol-
uble alpha-synuclein, indicating neuropathological
similarities between A30P disease patients and id-
iopathic PD, with a more severe neuropathology
in A30P carriers.
ANN NEUROL 2010;67:684689
The identification of the SNCA gene on chromosome
4q21-23 encoding alpha-synuclein as the first gene
responsible for autosomal dominantly inherited Parkinson
disease (PD) and the subsequent characterization of 3
point mutations in this gene—A53T, E46K, and A30P—
revealed a close clinical relationship between familial PD
and idiopathic PD (IPD).
1–3
Together with familial forms
of PD due to multiplications of the SNCA gene, these 3
point mutations have attracted neuropathological interest,
because of hopes that insights into the pathomechanisms
of inherited PD could be helpful to unravel the unknown
pathogenesis of the frequent synucleinopathy IPD.
4
Contrasting the close clinical similarities, the neuro-
pathological relationship between familial PD caused by
mutations in the alpha-synuclein–encoding SNCA gene
and IPD is not well understood, because neuropatholog-
ical data on familial PD are limited.
2,5,6
To gain more
insight into the neuropathology of familial PD, we per-
formed the first pathoanatomical study of the brain of an
A30P mutation carrier in the SNCA gene.
Subjects and Methods
The 69-year-old male index patient from the German A30P
family received diagnosis of PD at the age of 54 years and was
successfully treated with L-dopa for the next 2 years.
7
The clin-
ical course until age 65 years included occurrences of L-dopa–
related complications (ie, hallucinations). With further disease
progression, the patient lost independence in all activities of
daily living and was unable to walk without assistance. During
the last years prior to his death, the patient also suffered from a
progressive cognitive decline. At the age of 69 years, he became
mutistic, dysphagic, and bedridden, and was kept alive by per-
cutaneous endoscopic gastrostomy (PEG). He died of respiratory
failure in a poor state of general health.
The brains of the A30P patient and 6 control individuals
(1 female, 5 males; mean age at death 60.7 12.8 years) were
examined in accordance with the Ethics Committee guidelines
of the Faculty of Medicine at Goethe University of Frankfurt
am Main. The brain of the A30P patient displayed a severe de-
pigmentation of the substantia nigra, as well as a slight atrophy
of the frontal, temporal, and parietal cerebral lobes (Fig 1B).
After fixation of the brains by immersion in 4% buffered form-
aldehyde solution, the cerebral, cerebellar, and brainstem tissue
blocks of the A30P patient and of 5 control cases were embed-
ded in polyethylene glycol (PEG 1000, Merck, Darmstadt, Ger-
many). For detailed tissue processing and staining procedures,
see the Supplemental Material.
The severity of neuronal loss and the frequency of alpha-
synuclein–immunopositive Lewy bodies (LBs), Lewy neurites
(LNs), and glial inclusions in the brain of the A30P index pa-
From the
1
Institute of Clinical Neuroanatomy, Dr Senckenberg Anat-
omy, Goethe University, Frankfurt am Main, Germany;
2
Center of
Neurology and Hertie-Institute for Clinical Brain Research, University
of Tu¨ bingen, Tu¨ bingen, Germany;
3
Department of Medical Genetics,
University of Tu¨ bingen, Tu¨ bingen, Germany;
4
Neuroanatomy and Mo-
lecular Brain Research, Ruhr-University Bochum, Bochum, Germany;
5
Department of Neuropathology, Heinrich-Heine-University, Du¨ ssel-
dorf, Germany;
6
Department of Neurology, Mayo Clinic, Jacksonville,
FL;
7
Neuropathology Laboratory, Mayo Clinic, Jacksonville, FL;
8
De-
partment of Human Physiology, Flinders University School of Medi-
cine, Bedford Park, Australia;
9
Institute of Brain Research, University of
Tu¨ bingen, Tu¨ bingen, Germany;
10
Institute for Cellular and Molecular
Anatomy, Goethe University, Frankfurt am Main, Germany; and
11
De-
partment of Pathology and Medical Biology, University Medical Cen-
ter Groningen, University of Groningen, Groningen, the Netherlands.
Address correspondence to Dr Ru¨ b, Institute of Clinical Neuroanat-
omy, Dr. Senckenberg Anatomy, Goethe University, Theodor-Stern-Kai
7, D-60590 Frankfurt/Main, Germany; E-mail: Drueb@gmx.de or Dr
Kru¨ ger, Center of Neurology and Hertie-Insitute for Clinical Brain Re-
search, University of Tu¨ bingen, Hoppe-Seyler-Str. 3, 72076 Tu¨ bingen,
Germany; E-mail: rejko.krueger@uni-tuebingen.de
Received Jun 30, 2009, and in revised form Dec 10. Accepted for
publication Dec 22, 2009.
Published online in Wiley InterScience (www.interscience.wiley.com).
DOI: 10.1002/ana.21966
Additional Supporting Information may be found in the online version
of this article.
ANNALS of Neurology
684 Volume 67, No. 5
tient were semiquantitatively assessed (not discernible, ; slight,
; marked, ⫹⫹; severe, ⫹⫹⫹; Supplementary Tables 1–5).
Subcellular fractionation of frontal or entorhinal cortex
tissue from the A30P patient, 1 patient from a Greek-American
kindred carrying the A53T mutation in the SNCA gene,
8
1 pa-
tient with dementia with Lewy bodies (DLB), 2 IPD patients,
and 2 control individuals (Supplementary Table 6) was per-
formed as described previously.
9,10
Western blots using 3 differ-
ent antibodies against alpha-synuclein (N-terminus: PA1-38705,
Affinity Bioreagents, Golden, CO; NAC domain: Mc42, Trans-
labs, UK; C-terminus:15G7; AG Scientific, San Diego, CA)
were reproduced 3 times with similar results, with each sample
loaded at least 2 times on different blots.
Electron microscopy was performed to elucidate the ultra-
structure of neuronal alpha-synuclein aggregates in the substan-
tia nigra, basal nucleus of Meynert, and dorsal motor vagal nu-
cleus (DMV) of the A30P patient and 1 patient with IPD as
control (female; age at death, 76 years; disease duration, 26
years; Braak stage 5) as described previously.
11
Results
Investigation of pigment-Nissl stained tissue sections re-
vealed neuronal loss in the pars compacta of the substan-
tia nigra (see Fig 1D, E), locus coeruleus, and DMV (see
Fig 1F, G).
LBs and LB-like inclusions occurred as spherical,
reniform, or globose neuronal inclusions with smooth sur-
faces (Fig 2A). LNs had a club-or corkscrew-shaped ap-
pearance, or were short and stubby or slender, elongated,
and thread-like (see Fig 2A). Alpha-synuclein–immu-
nopositive LBs were present in all areas of the cerebral
cortex (neocortex: predominantly in layers V and VI;
ento-and transentorhinal regions: predominantly in layers
II, III, V, and VI; subiculum, presubiculum, hippocampal
CA1, CA2, and CA3 sectors: predominantly in pyramidal
layers) (Supplementary Fig 1A, B), in all nuclei of the
basal forebrain, basal ganglia, amygdala, and hypothala-
Š
FIGURE: 1 Macroscopic brain aspects in A30P and A30P
mutation-related nerve cell loss. (A) Lateral aspect of the
left cerebral hemisphere of a 53-year-old male individual
without a prior medical history of neurological or psychi-
atric disease. (B) Lateral aspect of the left cerebral hemi-
sphere from the A30P index patient. Note the widened
sulci of the frontal (arrowheads), temporal (asterisk), and
parietal lobes (arrow). (C) Pedigree of the German A30P
family indicating the deceased index patient (black arrow;
filled black symbols: affected family members according to
the United Kingdom Parkinson’s Disease Brain Bank crite-
ria; filled gray symbols: individuals displaying subtle extra-
pyramidal symptoms; circles: females; squares: males;
oblique slashes: deceased individuals; open symbols:
healthy family members). (D) Frontal section through the
rostral midbrain of a typical 59-year-old male control case
with the dorsal portion of the substantia nigra (SN). (E)
Severely degenerated SN of the A30P index patient. (F)
Horizontal section through the mid portion of the medulla
oblongata of an 84-year-old male control case showing the
dorsal motor vagal nucleus (DMV). (G) Markedly degener-
ated DMV of the A30P index patient. Arrows point to
surviving nerve cells. A–D: aldehyde fuchsin-Darrow red
staining, 100
m percutaneous endoscopic gastrostomy
sections. I first generation; II second generation; III
third generation; IV fourth generation; V fifth gener-
ation; PP peripeduncular nucleus; RD red nucleus;
CP cerebral peduncle; SOL solitary tract; IC inter-
calate nucleus; XII hypoglossal nucleus.
Seidel et al: A30P Family Pathoanatomy
May, 2010 685
mus (see Supplementary Fig 1C, D), in select regions of
the thalamus (see Supplementary Fig 1E–H), subthalamus
(Supplementary Fig 2A), and brainstem (see Supplemen-
tary Fig 2B–F), and in the cerebellum.
In the thalamus, basal nucleus of Meynert, substan-
tia nigra, DMV, gigantocellular reticular nucleus, raphes
magnus nucleus, and intermediate reticular zone, we ob-
served alpha-synuclein–immunopositive neurons without
FIGURE: 2 Alpha-synuclein immunocytochemical and electron microscopic findings in the A30P index patient. (A) Lewy
bodies (LBs) (large arrows) and Lewy neurites (arrowheads) in the dorsal motor vagal nucleus. Small arrows point to coiled
bodies. (B) Gleason score glial fibrillary acidic protein (GFAP)-immunopositive astrocytes in the transentorhinal cortex con-
taining alpha-synuclein immunopositive deposits (arrowheads). (C) Substantia nigra: Typical coiled-up alpha-synuclein immu-
nopositive oligodendroglial inclusion (brown) surrounding the nucleus (blue) of the affected oligodendrocyte. (D) The dorsal
motor vagal nucleus of the A30P patient exhibits various LBs (arrows) with a dark inner core and a ring-like light outer zone.
(E, F) Electron micrograph of the LB in the lower left corner of D (asterisk). The LB shows a dense granular core (Co),
surrounded by a lighter outer zone (OZ), which contains loosely arranged filaments (F). (G, H) Electron micrograph of an LB
found in an idiopathic Parkinson disease case. Note the high structural similarity of the LBs from both cases. (A) Anti–
alpha-synuclein immunocytochemistry; (B) anti-GFAP (3,3-diaminobenzidine [DAB], brown)/anti–alpha-synuclein (SK4700,
blue-gray) double immunostaining; (C) antitransferrin (SK4700, blue-gray)/anti–alpha-synuclein (DAB, brown) double immu-
nostaining; (A–C) 100
m percutaneous endoscopic gastrostomy sections; (D) toluidine blue-stained semithin section, 0.75
m;
(E–H) uranyl acetate and lead citrate contrasted ultrathin section, 100nm).
ANNALS of Neurology
686 Volume 67, No. 5
LBs or LNs. These neurons, however, were characterized
by the presence of fine alpha-synuclein–immunopositive,
slightly brownish, and loosely scattered cytoplasmic gran-
ules sparing the nucleus and often extending into the pro-
cesses of nerve cells. In addition, alpha-synuclein immu-
nopositivity and/or LB-like inclusions were observed in
central nervous white matter components (see Supple-
mentary Fig 2E–H).
Alpha-synuclein–immunopositive coiled bodies (see
Fig 2A, C; Supplementary Fig 2A, B) and glial fibrillary
acidic protein-immunopositive astrocytes were present in
all gray and white matter components of the telencepha-
lon, diencephalon, brainstem, and cerebellum of the
A30P patient. Anti–alpha-synuclein/antitransferrin double
immunostaining confirmed the purely oligodendroglial lo-
calization of the coiled bodies, and AT8 and PHF-1 im-
munostaining excluded their tau immunoreactivity (see
Fig 2C). Alpha-synuclein–immunopositive and tau-
negative astrocytes (see Fig 2B) occurred in all nuclei of
the amygdala and the septum, striatum, claustrum, select
thalamic nuclei (ie, central lateral nucleus, limitans-
suprageniculate complex), temporal neocortex, and ento-
and transentorhinal regions. No colocalization of tau with
alpha-synuclein was observed in neurons and oligoden-
drocytes of the dorsal raphe nucleus and the intermediate
reticular zone (Supplementary Fig 3).
The Alzheimer disease (AD)-related cortical cy-
toskeletal pathology as assessed by AT8 and PHF-1 im-
munostaining corresponded to Braak stage II, and the
brain -amyloidosis to phase 1 of the schema proposed by
Thal and colleagues.
12,13
Ultrastructural studies confirmed neuronal loss in
the substantia nigra and DMV nucleus. The majority of
remaining nigral neurons underwent dark cell degenera-
tion. Several surviving nigral neurons displayed round so-
matic inclusions with a denser center and a lighter outer
zone as a correlate of LBs. In the DMV nucleus, LBs were
detected with a dark dense core (see Fig 2D) and a broad
lighter outer zone reflecting the classical brainstem type of
LB.
14
They were frequently localized in the cytoplasm of
nerve cells, but also occurred in the neuropil, suggesting
dendritic or axonal localization. Electron microscopy re-
FIGURE: 3 Western blots of sequentially extracted alpha-
synuclein. (A) Frontal cortex tissues from 2 healthy control
individuals, 1 patient with dementia with Lewy bodies
(DLB), 2 idiopathic Parkinson disease (IPD) patients (Braak
stage 3 and 5), the A30P patient, and a patient carrying
the A53T mutation were sequentially extracted with Tris-
HCl (TBS) and urea (U; insoluble fraction) followed by im-
munoblotting with antibodies specific to the epitopes indi-
cated in A. The IPD patient with advanced Braak stage 5,
the DLB patient, and both mutation carriers revealed in-
soluble monomeric (
-SYN)
1
and oligomeric alpha-synuclein
(
-SYN)
n
species in the urea fraction, which were most pro-
nounced in the A30P patient (asterisks). (B) Sequential pro-
tein extraction of the entorhinal and frontal cortices of the
A30P patient and an A53T patient revealed a strong alpha-
synuclein signal in the urea-soluble fractions of the A30P
mutation carrier (asterisks). (C) Schematic representation of
the human alpha-synuclein protein and epitopes of the an-
tibodies used in this study. T/T; Triton X-100-soluble, mem-
brane bound fraction.
Seidel et al: A30P Family Pathoanatomy
May, 2010 687
vealed a dense dark granular center and a ring-like outer
zone with abundant fibrils (see Fig 2E, F), similar to the
ultrastructural characteristics of LBs from an IPD patient
(Braak stage 5; see Fig 2G, H).
Biochemical sequential extraction of alpha-synuclein
of frontal cortex of controls, DLB patients, IPD patients
(Braak stage 3 and stage 5), the A30P patient, and an
A53T patient revealed soluble monomeric alpha-synuclein
in all samples (Tris-HCl; Fig. 3A). The presence of mo-
nomeric and oligomeric alpha-synuclein species in the
urea fraction was not observed in controls and disclosed
insolubility of alpha-synuclein that was most prominent
in the A30P case and DLB patient (see Fig 3A). A30P
alpha-synuclein insolubility was prominent in both the
entorhinal and frontal cortex of the A30P patient substan-
tiating immunohistochemically determined LB load in
these 2 brain areas (see Fig 3B).
Discussion
Our study provides first insights into the pathoanatomy
of familial PD caused by the A30P mutation in the SNCA
gene and demonstrates a widespread central nervous oc-
currence of alpha-synuclein–immunopositive inclusions
and the advantage of investigations of complete tissue se-
ries for the identification of more subtle pathological al-
terations.
15,16
Application of sensitive immunostaining methods
revealed tau-immunoreactive cortical cytoskeletal pathol-
ogy in the index patient. Because no additional tau-
positive neuronal and glial inclusions associated with
other known human tauopathies were observed, the cor-
tical tau pathology most likely corresponded to the early
Braak AD stage II.
12
In addition, alpha-synuclein immu-
nocytochemistry in the A30P patient did not reveal fea-
tures of neuronal inclusions or the distribution pattern of
alpha-synuclein–immunopositive oligodendroglial inclu-
sions typical for multiple system atrophy.
15
As in IPD, the A30P patient’s brain showed neuro-
nal loss in the substantia nigra, DMV, and locus coer-
uleus and displayed recognized neuronal pathologies (ie,
LBs, LNs, and LB-like inclusions) in brain structures typ-
ically affected in IPD (eg, neo-and allocortex, thalamus,
cerebellum, substantia nigra, pedunculopontine nucleus,
DMV), as well as in brainstem nuclei and central nervous
fiber tracts that are not or are only less severely affected in
IPD (eg, rostral interstitial nucleus of the medial longitu-
dinal fascicle, reticulotegmental nucleus of the pons, me-
dial vestibular nucleus, spinocerebellar tracts, and cerebel-
lar peduncles).
17,18
In addition, as in IPD, alpha-
synuclein–immunopositive astroglial and oligodendroglial
inclusions were present in the A30P patient.
19
Among the currently known brain pathologies re-
ported in familial PD forms caused by mutations or mul-
tiplications in the SNCA gene, the brain pathology of the
A30P index patient most closely resembled the brain al-
terations observed in IPD individuals.
2,5,6,20
Neuropatho-
logical discrepancies that strengthen the pathogenic role
of A30P mutant alpha-synuclein and may indicate differ-
ent etiopathogenic mechanisms included: (1) substantial
and more severe load of alpha-synuclein aggregates in the
cerebellum and precerebellar and occulomotor brainstem
nuclei than in IPD; and (2) presence of more glial aggre-
gates, named coiled bodies, in the A30P brain than in
IPD. The presence of concurrent tau pathology in A53T
mutation carriers or the accumulation of alpha-synuclein
and tau within the same neuronal aggregates in carriers of
alpha-synuclein multiplications underscore important dis-
crepancies between IPD patients and the A30P mutation
carrier studied here.
5,20
A pathological relationship between the alpha-
synuclein pathology of A30P and IPD is also supported
(1) by our ultrastructural analyses of LBs in the A30P
mutation carrier in comparison to an IPD patient, and
(2) by our biochemical findings, which showed insolubil-
ity of alpha-synuclein in the A30P patient and in IPD
patients. This insolubility reflects a substantial aggregate-
forming capacity of A30P mutant alpha-synuclein and
contrasts with previous in vitro observations on fibril for-
mation of different mutant alpha-synuclein species.
21
In summary, the pathological similarities between
the A30P patient and IPD strongly support the view that
familial PD caused by the A30P mutation in the SNCA
gene not only is closely related to IPD clinically, but also
pathologically. Although additional postmortem A30P
studies are required to ultimately define this neuropatho-
logical relationship, our study provides major implications
for the validation of transgenic animal models for PD.
Acknowledgment
This study was supported by the Deutsche Forschungsge-
meinschaft (RU 1215/1-2, KR2119/3-1) and the Federal
Ministry for Education and Research (01GS01834, R.K.
and O.R.).
We thank the A30P family for their continued sup-
port and interest in this project; P. Davies for the dona-
tion of the PHF-1 antibody; M. Babl, B. Meseck-
Selchow, M. Bouzrou for processing of tissue sections and
immunocytochemistry; M. Lo¨bbecke-Schuhmacher for
electron microscopy; and M. Hu¨tten for secretarial assis-
tance.
ANNALS of Neurology
688 Volume 67, No. 5
Potential Conflicts of Interest
Udo Ru¨b is the Executive Editor of Neuropathology and
Applied Neuropathology.
References
1. Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the
alpha-synuclein gene identified in families with Parkinson’s dis-
ease. Science 1997;276:2045–2047.
2. Zarranz JJ, Alegre J, Gomez-Estaban JC, et al. The new muta-
tion, E46K, of alpha-synuclein causes Parkinson and Lewy body
dementia. Ann Neurol 2004;55:164–173.
3. Kru¨ ger R, Kuhn W, Mu¨ ller T, et al. Ala30Pro mutation in the
gene encoding alpha-synuclein in Parkinson’s disease. Nat Genet
1998;18:106–108.
4. Schiesling C, Kieper N, Seidel K, Kru¨ ger R. Familial PD-genetics,
clinical phenotype and neuropathology in relation to the com-
mon sporadic form of the disease. Neuropathol Appl Neurobiol
2008;34:255–271.
5. Duda JE, Giasson BI, Mabon ME, et al. Concurrence of alpha-
synuclein and tau brain pathology in the Contursi kindred. Acta
Neuropathol 2002;104:7–11.
6. Markopoulou K, Dickson DW, McComb RD, et al. Clinical, neu-
ropathological and genotypic variability in SNCA A53T familial
Parkinson’s disease: variability in familial Parkinson’s disease.
Acta Neuropathol 2008;116:25–35.
7. Kru¨ ger R, Kuhn W, Leenders KL, et al. Familial parkinsonism with
synuclein pathology: clinical and PET studies of A30P mutation
carriers. Neurology 2001;56:1355–1362.
8. Markopoulou K, Wszolek ZK, Pfeiffer RF. A Greek-American kin-
dred with autosomal dominant, levodopa-responsive parkinson-
ism and anticipation. Ann Neurol 1995;38:373–378.
9. Braak H, Del Tredici K, Ru¨ b U, et al. Staging of brain pathology
related to sporadic Parkinson’s disease. Acta Neuropathol 2003;
24:197–211.
10. Tofaris GK, Razzaq A, Ghetti B, et al. Ubiquitination of alpha-
synuclein in Lewy bodies is a pathological event not associated
with impairment of proteasome function. J Biol Chem 2003;278:
44405–44411.
11. Petrasch-Parwez A, Nguyen HP, Lo¨bbecke-Schumacher M, et al.
Cellular and subcellular localization of Huntingtin aggregates in
the brain of a rat transgenic for Huntington disease. J Comp
Neurol 2007;501:716–730.
12. Braak H, Braak E. Neuropathological staging of Alzheimer-related
changes. Acta Neuropathol 1991;82:239–259.
13. Thal DR, Ru¨b U, Orantes M, Braak H. Phases of Abeta-deposition
in the human brain and its relevance for the development of AD.
Neurology 2002;58:1791–1800.
14. Kosaka K. Lewy bodies in cerebral cortex, report of three cases.
Acta Neuropathol 1978;42:127–134.
15. Braak H, Ru¨b U, Del Tredici K. Involvement of precerebellar nu-
clei in multiple system atrophy. Neuropathol Appl Neurobiol
2003;29:60–76.
16. Ru¨b U, Brunt ER, Del Turco D, et al. Guidelines for the patho-
anatomical examination of the lower brain stem in ingestive and
swallowing disorders and its application to a dysphagic spino-
cerebellar ataxia type 3 patient. Neuropathol Appl Neurobiol
2003;29:1–13.
17. Braak H, Del Tredici K. Neuroanatomy and pathology of sporadic
Parkinson’s disease. Berlin, Heidelberg: Springer, 2009.
18. Ru¨b U, Del Tredici K, Schultz C, et al. Parkinson’s disease: the
thalamic components of the limbic loop are severely impaired by
alpha-synuclein immunopositive inclusion body pathology. Neu-
robiol Aging 2002;23:245–254.
19. Wakabayashi K, Hayashi S, Yoshimoto M, et al. NACP/alpha-
synuclein-positive filamentous inclusions in astrocytes and oligo-
dendrocytes of Parkinson’s disease brains. Acta Neuropathol
2000;99:14–20.
20. Gwinn-Hardy K, Mehta ND, Farrer M, et al. Distinctive neuropa-
thology revealed by alpha-synuclein antibodies in hereditary par-
kinsonism and dementia linked to chromosome 4p. Acta Neuro-
pathol 2000;99:663–672.
21. Conway KA, Harper JD, Lansbury PT. Accelerated in vitro fibril
formation by a mutant alpha-synuclein linked to early-onset Par-
kinson disease. Nat Med 1998;4:1318–1320.
Endogenous Neurosteroid
Synthesis Modulates Seizure
Frequency
Courtney Lawrence, Brandon Scott Martin,
Chengsan Sun, John Williamson, and
Jaideep Kapur, PhD
Inhibitory neurosteroids, molecules generated in glia
from circulating steroid hormones and de novo from
cholesterol, keep seizures in check in epileptic ani-
mals. They can enhance inhibitory transmission medi-
ated by gamma-aminobutyric acid receptors and have
anticonvulsant action.
ANN NEUROL 2010;67:689693
Many pathological changes in the epileptic brain in-
crease the propensity for seizures; however, certain
biological mechanisms and molecules appear to keep sei-
zures in check in epileptic animals. Inhibitory neuros-
teroids, molecules generated in glia from circulating ste-
roid hormones and de novo from cholesterol, are
candidate molecules for checking seizures because they
can enhance inhibitory transmission mediated by gamma-
aminobutyric acid (GABA)
A
receptors and have anticon-
vulsant action.
1
Neurosteroids modulate both synaptic
and tonic inhibition mediated by GABA
A
receptors.
2,3
Anticonvulsant action of naturally occurring neuro-
steroids was demonstrated soon after their GABA
A
recep-
tor action was found.
4
The potency of anticonvulsant ac-
From the Department of Neurology, University of Virginia, Charlottes-
ville, Virginia.
Address correspondence to Dr Kapur, Department of Neurology, Box
800394, University of Virginia-HSC, Charlottesville, VA 22908. E-mail:
jk8t@virginia.edu
Received Jun 18, 2009, and in revised form Jan 3, 2010. Accepted for
publication Jan 22, 2010.
Published in Wiley InterScience (www.interscience.wiley.com). DOI:
10.1002/ana.21989
Lawrence et al: Endogenous Neurosteroids
May, 2010 689
... Some autosomal-dominantly hereditary familial PD lineages were found to have a point genetic mutation coding the presynaptic α-synuclein protein [11,48]. The pathological similarity between the A30P patient and Idiopathic Parkinson's disease (IPD) clearly support the hypothesis that familial PD induced by the mutation (A30P) in the gene of α-synuclein is not only clinically but also pathologically connected to IPD [14]. ...
Article
Full-text available
Background Alpha synuclein (α-synuclein) is coded by SNCA gene and found in a helical form with phospholipids or in an unfolded arrangement in the cytosol and belongs to the synuclein family other than beta synuclein and gamma synuclein. It is a short protein made of 140 amino acids with three domains: an N-terminal lipid binding helix, a non-amyloid-ß component (NAC), and an acidic tail at the C-terminus. α-Synuclein is present in aggregated and fibrillar form in Lewy bodies and its association has been related to multiple system atrophy (MSA), Parkinson’s disease (PD), and Dementia with Lewy bodies (DLB). Our objective is to investigate and prioritise the possible nsSNPs in the α-synuclein protein that have been potentially connected to human neurodegenerative diseases. Results We used the series of computational tools to predict the mutation's harmful effect on three-dimensional structure of α-synuclein based on consensus approach. Our findings pointed to a significant computational blueprint for discovering nsSNPs connected to neurodegenerative illnesses from a large SNP data set while also minimising the expenses of experimentally showing harmful nsSNPs. Conclusions The prioritised G25S (rs1433622151), V66E (rs1261243630), and V77D (rs745815563) mutations can be employed in additional experimental studies to assess the α-synuclein protein mutation in relation to neurodegenerative illnesses and develop a therapeutics against them.
... A family from Germany had typical PD associated with a A30P mutation; 24 the first pathological appraisal of these patients showed similar lesions to those described above for the A53T patients. 25 Another large family from Spain shows a mixture of phenotypes from typical PD to the more extensive Lewy body disease. These patients have the E46K mutation. ...
Chapter
In recent years, medical developments have resulted in an increase in human life expectancy. Some developed countries now have a larger population of individuals aged over 64 than those under 14. One consequence of the ageing population is a higher incidence of certain neurodegenerative disorders. In order to prevent these, we need to learn more about them. This book provides up-to-date information on the use of transgenic mouse models in the study of neurodegenerative disorders such as Alzheimer's and Huntington's disease. By reproducing some of the pathological aspects of the diseases, these studies could reveal the mechanism for their onset or development. Some of the transgenic mice can also be used as targets for testing new compounds with the potential to prevent or combat these disorders. The editors have extensive knowledge and experience in this field and the book is aimed at undergraduates, postgraduates and academics. The chapters cover disorders including: Alzheimer's disease, Parkinson's disease, Huntington's and other CAG diseases, amyotrophic lateral sclerosis (ALS), recessive ataxias, disease caused by prions, and ischemia.
... Although genetic mutations in PD are rare and represent only about 10% of all PD cases, animal models carrying these mutations are important as they represent potential therapeutic targets. Moreover, in the case of αSyn, genome-wide association studies have linked variations in the SNCA gene to a higher risk of PD [454,455], and aggregation of αSyn is also present in sporadic PD [10]. ...
Thesis
Age-related neurodegenerative disorders such as Parkinson’s disease (PD) represent a challenge to contemporary society. The increase in life expectancy and the lack of therapies addressing the disease cause make these disorders a high burden to patients, caretakers and society in general. Aggregation of αSynuclein (αSyn) and presence of characteristic motor symptoms are cardinal features of PD. The onset of PD motor symptoms is however preceded by non-motor symptoms that can start decades earlier. These may be associated with initiation of αSyn pathology in the peripheral nervous system, more precisely in the enteric nervous system (ENS) and/or olfactory system. Several studies have been performed in patients as well as in animal models to investigate the spreading of αSyn from the periphery to the central nervous system (CNS). Animal models of αSyn gut-to-brain spreading are usually based on the injection of αSyn aggregates or on viral overexpression of αSyn, with both cases implying external manipulation. Hence, the main aim of this thesis was to characterise a novel, non-invasive, transgenic mouse model of spontaneous αSyn gut-to-brain spreading.This mouse model, named Vitras, expresses aggregation prone human 1-120αSyn exclusively in the gut in a αSyn-null background. To determine whether the presence of human or mouse αSyn in the background could affect the spreading of αSyn, two additional versions of the Vitras line, expressing either endogenous mouse αSyn (Vitras6J) or full-length human αSyn (VitrasBAC), were characterised. We found that gut-to-brain αSyn spreading can occur independently from the presence of endogenous full-length αSyn and was present at 3 months of age. The first gut-derived αSyn-positive neurons in the brain were detected in the dorsal motor nucleus of the vagus nerve, followed by other brainstem nuclei. In addition to the brain, gut-derived αSyn was also found in the spinal cord, lungs and heart. Although none of the mouse lines presented constipation, Vitras mice developed a hyperactive behaviour at 12 months of age. To understand the mechanisms of αSyn spreading at a cellular level, a neuronal co-culture system of ENS-CNS neurons was set up. Moreover, in an attempt to identify compounds to target αSyn aggregation a new oligomer-modulator, Sery315b, was tested in various cellular models. Due to time limitations these studies were not completed. In conclusion, the work demonstrated that the Vitras models are valuable tools to investigate the mechanisms of αSyn pathology spreading from the ENS to the CNS and to test possible therapies aimed at slowing down disease progression in PD.
... The first gene mutation identified in patients with PD was associated with the Ala53 residue (Polymeropoulos et al., 1997). Since then, other mutations responsible for the onset of PD have been identified as associated with the Ala30 (Seidel et al., 2010), Glu46 (Zarranz et al., 2004), His50 (Appel-Cresswell et al., 2013 and Gly51 (Lesage et al., 2013) residues. Modified from Ottolini, D., Cali, T., Szabo, I., Brini, M., 2017. ...
Chapter
Full-text available
Alpha-synuclein (α-synuclein) is a small, acidic protein containing 140 amino acids, highly expressed in the brain and primarily localized in the presynaptic terminals. It is found in high concentrations in Lewy Bodies, proteinaceous aggregates that constitute a typical histopathologic hallmark of Parkinson's disease. Altered environmental conditions, genetic mutations and post-translational changes can trigger abnormal aggregation processes with the increased frequency of oligomers, protofibrils, and fibrils formation that perturbs the neuronal homeostasis leading to cell death. Relevant to neuronal activity, a function of α-synuclein that has been extensively detailed is its regulatory actions in the trafficking of synaptic vesicles, including the processes of exocytosis, endocytosis and neurotransmitter release. Most recently, increasing attention has been paid to the possible role that α-synuclein plays at a postsynaptic level by interacting with selective subunits of the glutamate N-methyl-d-aspartate receptor, altering the corticostriatal plasticity of distinct neuronal populations.
Article
Full-text available
Background Cutaneous phosphorylated alpha-synuclein (p-α-syn) deposition is an important biomarker of idiopathic Parkinson’s disease (iPD). Recent studies have reported synucleinopathies in patients with common genetic forms of PD. Objective This study aimed to detect p-α-syn deposition characteristic in rare genetic PD patients with CHCHD2 or RAB39B mutations. Moreover, this study also aimed to describe peripheral alpha-synuclein prion-like activity in genetic PD patients, and acquire whether the cutaneous synucleinopathy characteristics of genetic PD are consistent with central neuropathologies. Methods We performed four skin biopsy samples from the distal leg (DL) and proximal neck (C7) of 161 participants, including four patients with CHCHD2 mutations, two patients with RAB39B mutations, 16 patients with PRKN mutations, 14 patients with LRRK2 mutations, five patients with GBA mutations, 100 iPD patients, and 20 healthy controls. We detected cutaneous synucleinopathies using immunofluorescence staining and a seeding amplification assay (SAA). A systematic literature review was also conducted, involving 64 skin biopsies and 205 autopsies of genetic PD patients with synucleinopathy. Results P-α-syn was deposited in the peripheral cutaneous nerves of PD patients with CHCHD2, LRRK2, or GBA mutations but not in those with RAB39B or PRKN mutations. There were no significant differences in the location or rate of α-syn-positive deposits between genetic PD and iPD patients. Peripheral cutaneous synucleinopathy appears to well represent brain synucleinopathy of genetic PD, especially autosomal dominant PD (AD-PD). Cutaneous α-synuclein SAA analysis of iPD and LRRK2 and GBA mutation patients revealed prion-like activity. Conclusion P-α-syn deposition in peripheral cutaneous nerves, detected using SAA and immunofluorescence staining, may serve as an accurate biomarker for genetic PD and iPD in the future.
Article
Full-text available
Parkinson’s disease (PD) is primarily idiopathic and a highly heterogenous neurodegenerative disease with patients experiencing a wide array of motor and non-motor symptoms. A major challenge for understanding susceptibility to PD is to determine the genetic and environmental factors that influence the mechanisms underlying the variations in disease-associated traits. The pathological hallmark of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta region of the brain and post-mortem Lewy pathology, which leads to the loss of projecting axons innervating the striatum and to impaired motor and cognitive functions. While the cause of PD is still largely unknown, genome-wide association studies provide evidence that numerous polymorphic variants in various genes contribute to sporadic PD, and 10 to 15% of all cases are linked to some form of hereditary mutations, either autosomal dominant or recessive. Among the most common mutations observed in PD patients are in the genes LRRK2, SNCA, GBA1, PINK1, PRKN, and PARK7/DJ-1. In this review, we cover these PD-related mutations, the use of induced pluripotent stem cells as a disease in a dish model, and genetic animal models to better understand the diversity in the pathogenesis and long-term outcomes seen in PD patients.
Article
Full-text available
Antisense oligonucleotides (ASOs) are single-stranded nucleic acid strings that can be used to selectively modify protein synthesis by binding complementary (pre-)mRNA sequences. By specific arrangements of DNA and RNA into a chain of nucleic acids and additional modifications of the backbone, sugar, and base, the specificity and functionality of the designed ASOs can be adjusted. Thereby cellular uptake, toxicity, and nuclease resistance, as well as binding affinity and specificity to its target (pre-)mRNA, can be modified. Several neurodegenerative diseases are caused by autosomal dominant toxic gain-of-function mutations, which lead to toxic protein products driving disease progression. ASOs targeting such mutations—or even more comprehensively, associated variants, such as single nucleotide polymorphisms (SNPs)—promise a selective degradation of the mutant (pre-)mRNA while sparing the wild type allele. By this approach, protein expression from the wild type strand is preserved, and side effects from an unselective knockdown of both alleles can be prevented. This makes allele-specific targeting strategies a focus for future personalized therapies. Here, we provide an overview of current strategies to develop personalized, allele-specific ASO therapies for the treatment of neurodegenerative diseases, such Huntington’s disease (HD) and spinocerebellar ataxia type 3 (SCA3/MJD).
Article
Full-text available
Neurons are post-mitotic cells that allocate huge amounts of energy to the synthesis of new organelles and molecules, neurotransmission and to the maintenance of redox homeostasis. In neurons, autophagy is not only crucial to ensure organelle renewal but it is also essential to balance nutritional needs through the mobilization of internal energy stores. A delicate crosstalk between the pathways that sense nutritional status of the cell and the autophagic processes to recycle organelles and macronutrients is fundamental to guarantee the proper functioning of the neuron in times of energy scarcity. This review provides a detailed overview of the pathways and processes involved in the balance of cellular energy mediated by autophagy, which when defective, precipitate the neurodegenerative cascade of Parkinson's disease, frontotemporal dementia, amyotrophic lateral sclerosis or Alzheimer's disease.
Article
Epidemiology is the study of the distribution of disease in human populations, which is important in evaluating burden of illness, identifying modifiable risk factors, and planning for current and projected needs of the health care system. Parkinson's disease (PD) is the second most common serious neurodegenerative illness and is expected to further increase in prevalence. Cognitive changes are increasingly viewed as an integral non-motor feature in PD, emerging even in the prodromal phase of the disease. The prevalence of PD-MCI ranges from 20% to 40% depending on the population studied. The incidence of PD-dementia increases with duration of disease, with estimates growing from 3% to 30% of individuals followed for 5 years or less to over 80% after 20 years. There are several challenges in estimating the frequency of cognitive change, including only recently standardized diagnostic criteria, variation depending on exact neuropsychological evaluations performed, and differences in population sampling. Clinical features associated with cognitive decline include older age, increased disease duration and severity, early gait dysfunction, dysautonomia, hallucinations and other neuropsychiatric features, the presence of REM behavior disorder, and posterior predominant dysfunction on neuropsychological testing. There is increasing evidence that genetic risk factors, in particular GBA and MAPT mutations, contribute to cognitive change. Possible protective factors include higher cognitive reserve and regular exercise. Important sequelae of cognitive decline in PD include higher caregiver burden, decreased functional status, and increased risk of institutionalization and mortality. Many remaining uncertainties regarding the epidemiology of cognitive change in PD require future research, with improved biomarkers and more sensitive and convenient outcome measures.
Article
Full-text available
Parkinson's disease (PD) is a common neurodegenerative disorder with a lifetime incidence of approximately 2 percent. A pattern of familial aggregation has been documented for the disorder, and it was recently reported that a PD susceptibility gene in a large Italian kindred is located on the long arm of human chromosome 4. A mutation was identified in the alpha-synuclein gene, which codes for a presynaptic protein thought to be involved in neuronal plasticity, in the Italian kindred and in three unrelated families of Greek origin with autosomal dominant inheritance for the PD phenotype. This finding of a specific molecular alteration associated with PD will facilitate the detailed understanding of the pathophysiology of the disorder.
Book
The proteinopathy sporadic Parkinson’s disease (sPD) is the second most frequent degenerative disorder of the human nervous system after Alzheimer’s disease. The a -synuclein inclusion body pathology (Lewy pathology) associated with sPD is distributed throughout the central, peripheral, and enteric nervous systems. The resulting nonrandom neuronal dysfunction and, in some regions, neuronal loss is reflected by a distinctive topographic distribution pattern of the Lewy pathology that, in the brain, has been staged. Except for olfactory structures and spinal cord constituents of the pain system, sensory components of the nervous system remain uninvolved or virtually intact. The most disease-related damage revolves around motor areas – particularly around superordinate centers of the limbic and visceromotor systems as well as portions of the somatomotor system. Vulnerable regions are interconnected anatomically and susceptible nerve cell types are not neurotransmitter-dependent. Not all clinical symptoms emerging in the course of sPD can be explained by a lack of dopamine in the nigrostriatal system. These include autonomic dysfunction, pain, hyp- or anosmia, excessive daytime sleepiness, REM sleep behavioral disorder, depression, anxiety, cognitive decline, and dementia. Against the background of the normal morphology and anatomy, the authors analyze the pathoanatomy of sPD in the nervous system at various neuropathological stages and summarize the potential functional consequences of the lesions.
Article
Eighty-three brains obtained at autopsy from nondemented and demented individuals were examined for extracellular amyloid deposits and intraneuronal neurofibrillary changes. The distribution pattern and packing density of amyloid deposits turned out to be of limited significance for differentiation of neuropathological stages. Neurofibrillary changes occurred in the form of neuritic plaques, neurofibrillary tangles and neuropil threads. The distribution of neuritic plaques varied widely not only within architectonic units but also from one individual to another. Neurofibrillary tangles and neuropil threads, in contrast, exhibited a characteristic distribution pattern permitting the differentiation of six stages. The first two stages were characterized by an either mild or severe alteration of the transentorhinal layer Pre-alpha (transentorhinal stages I-II). The two forms of limbic stages (stages III-IV) were marked by a conspicuous affection of layer Pre-alpha in both transentorhinal region and proper entorhinal cortex. In addition, there was mild involvement of the first Ammon's horn sector. The hallmark of the two isocortical stages (stages V-VI) was the destruction of virtually all isocortical association areas. The investigation showed that recognition of the six stages required qualitative evaluation of only a few key preparations.
Article
Previous genetic analysis of the familial Parkinson's disease Contursi kindred led to the identification of an Ala53Thr pathogenic mutation in the &#33-synuclein gene. We have re-examined one of the original brains from this kindred using new immunohistochemical reagents, thioflavin S staining and immunoelectron microscopy. Surprisingly, we uncovered a dense burden of &#33-synuclein neuritic pathology and rare Lewy bodies. Immunoelectron microscopy demonstrated fibrillar &#33-synuclein-immunoreactive aggregates. Unexpected tau neuritic and less frequent perikaryal inclusions were also observed. Some inclusions were comprised of both proteins with almost complete spatial disparity. We suggest that it is important to recognize that the neurodegenerative process caused by the Ala53Thr mutation in &#33-synuclein is not identical to that seen in typical idiopathic Parkinson's disease brains.
Article
The identification of the first gene in familial Parkinson's disease (PD) only 10 years ago was a major step in the understanding of the molecular mechanisms in neurodegeneration. Alpha-synuclein aggregation was not only recognized as a key event in neurodegeneration in patients carrying mutations in this gene, but it turned out to be the most consistent marker to define Lewy body pathology also in non-heritable idiopathic PD (IPD). Subsequent comprehensive pathoanatomical studies of IPD brains led to a novel concept of an ascending pathological process in variable stages that are reflected by alpha-synuclein aggregation at specific predilection sites. To date, more than seven genes are known to cause familial PD. The fact that these genetic forms of Parkinsonism present with clinical features indistinguishable from IPD, but may display neuropathological features that are not consistent with IPD, underscores the need of a more differentiated approach to familial and sporadic forms of Parkinsonism. Indeed, in distinct populations, mutations in one single gene were found to cause the disease in up to 40% of patients formerly described as ‘idiopathic’ cases. These findings indicate that IPD, as defined by a late-onset disorder with no (apparent) genetic contribution, is part of a clinical syndrome that becomes more and more heterogeneous in terms of aetiology, with overlapping clinical and pathoanatomical features. Thus in the present review, we discuss clues from familial PD to our understanding of the molecular pathogenesis of neurodegeneration with special consideration of the variable clinical and neuropathological aspects.
Article
The proteinopathy sporadic Parkinson's disease (sPD) is the second most frequent degenerative disorder of the human nervous system after Alzheimer's disease. The alpha-synuclein inclusion body pathology (Lewy pathology) associated with sPD is distributed throughout the central, peripheral, and enteric nervous systems. The resulting nonrandom neuronal dysfunction and, in some regions, neuronal loss is reflected in a topographic distribution pattern of the Lewy pathology that, in the brain, can be staged. Except for olfactory structures and spinal cord constituents of the pain system, sensory components of the nervous system remain uninvolved or virtually intact. The most disease-related damage revolves around motor areas--particularly around superordinate centers of the limbic and visceromotor systems as well as portions of the somatomotor system. Vulnerable regions are interconnected anatomically and susceptible nerve cell types are not neurotransmitter-dependent. Not all clinical symptoms emerging in the course of sPD can be explained by a lack of dopamine in the nigrostriatal system. These include autonomic dysfunction, pain, hyposmia or anosmia, excessive daytime sleepiness, rapid eye movement (REM) sleep behavioral disorder, depression, anxiety, cognitive decline, and dementia. Against the background of the normal morphology and anatomy, the authors analyze the pathoanatomy of sPD in the nervous system at various neuropathological stages and summarize the potential functional consequences of the lesions.
Article
The histochemical and structural properties and the topographical distribution pattern of Lewy bodies in the cerebral cortex as well as in the brain stem and diencephalon were studied in three cases. The Lewy bodies in the cerebral cortex were found in the small or medium-sized neurons of the fifth and sixth layers, particularly in the anterior frontal, temporal, insular, and cingulate cortex, and showed minor differences in their histochemical and structural properties from typical Lewy bodies in the brain stem and diencephalon. By light microscopy they were more irregular in shape, less eosinophilic, less sharply demarcated, and did not have clear halos and central cores. From the ultrastructural aspect, the filaments in them did not radiate, but were arranged at random, and circular profiles were not associated in the central zone. This type of Lewy body was also distributed in the basal ganglia. A close relationship between Lewy bodies and monoamines in the cerebral cortex of our cases was not recognized. These three cases showed also concomitant senile changes, i.e., senile plaques and neurofibrillary tangles.
Article
Eighty-three brains obtained at autopsy from nondemented and demented individuals were examined for extracellular amyloid deposits and intraneuronal neurofibrillary changes. The distribution pattern and packing density of amyloid deposits turned out to be of limited significance for differentiation of neuropathological stages. Neurofibrillary changes occurred in the form of neuritic plaques, neurofibrillary tangles and neuropil threads. The distribution of neuritic plaques varied widely not only within architectonic units but also from one individual to another. Neurofibrillary tangles and neuropil threads, in contrast, exhibited a characteristic distribution pattern permitting the differentiation of six stages. The first two stages were characterized by an either mild or severe alteration of the transentorhinal layer Pre-alpha (transentorhinal stages I-II). The two forms of limbic stages (stages III-IV) were marked by a conspicuous affection of layer Pre-alpha in both transentorhinal region and proper entorhinal cortex. In addition, there was mild involvement of the first Ammon's horn sector. The hallmark of the two isocortical stages (stages V-VI) was the destruction of virtually all isocortical association areas. The investigation showed that recognition of the six stages required qualitative evaluation of only a few key preparations.
Article
Interest is increasing concerning the role of genetic factors in the etiology of Parkinson's disease. We report the analysis of a Greek-American kindred with levodopa-responsive parkinsonism. Of the 98 individuals present in six generations of this pedigree, 16 individuals in three successive generations have developed parkinsonism. Affected members were examined both in Greece and in the United States. The clinical presentation consisted of asymmetric rigidity, resting tremor, bradykinesia, and postural instability, and symptoms were responsive to levodopa. The disease appears to be inherited in an autosomal dominant manner. The inheritance pattern and the development of parkinsonism in successive generations on two continents challenges environmental factors as the primary cause in the pathogenesis of parkinsonism in this kindred. Anticipation is present in this pedigree. The affected members in the third generation developed symptoms at ages 50 to 71, in the fourth at ages 40 to 55, and in the fifth at age 31 years. This is another example of a neurodegenerative disease with autosomal dominant inheritance and anticipation. A molecular genetic analysis of this pedigree is in progress.