ArticlePDF AvailableLiterature Review

Iron metabolism and its detection through MRI in parkinsonian disorders: a systematic review

Authors:

Abstract

Iron deposition in the brain normally increase with age, but its accumulation in certain regions is observed in a number of neurodegenerative diseases including Parkinson’s disease (PD) and other parkinsonisms. Whether iron overload leads to dopaminergic neuronal death in the SN of PD patients or is instead simply a by-product of the neurodegenerative progression is still yet to be ascertained. Magnetic resonance imaging (MRI) is a non-invasive method to assess brain iron content in PD patients. In PD, accurate radiologic visualization of basal ganglia is required. Deep gray matter nuclei are well presented in T2- and T2*-weighted images. T2*-weighted gradient-echo (GRE) is widely used to assess calcifications and also for iron detection. On the other hand, new methods specifically designed for detecting iron-induced susceptibility differences can be further improved by sequences like susceptibility-weighted imaging (SWI). In the present review, we aim to summarize the available data on brain iron deposition in PD.
REVIEW ARTICLE
Iron metabolism and its detection through MRI in parkinsonian
disorders: a systematic review
Sara Pietracupa
1
&Antonio Martin-Bastida
2
&Paola Piccini
2
Received: 10 April 2017 /Accepted: 22 August 2017
#Springer-Verlag Italia S.r.l. 2017
Abstract Iron deposition in the brain normally increase
with age, but its accumulation in certain regions is ob-
served in a number of neurodegenerative diseases in-
cluding Parkinsons disease (PD) and other parkinson-
isms. Whether iron overload leads to dopaminergic neu-
ronal death in the SN of PD patients or is instead sim-
ply a by-product of the neurodegenerative progression is
still yet to be ascertained. Magnetic resonance imaging
(MRI) is a non-invasive method to assess brain iron
content in PD patients. In PD, accurate radiologic visu-
alization of basal ganglia is required. Deep gray matter
nuclei are well presented in T2- and T2*-weighted im-
ages. T2*-weighted gradient-echo (GRE) is widely used
to assess calcifications and also for iron detection. On
the other hand, new methods specifically designed for
detecting iron-induced susceptibility differences can be
further improved by sequences like susceptibility-
weighted imaging (SWI). In the present review, we
aim to summarize the available data on brain iron de-
position in PD.
Keywords Parkinsonian disorders .Parkinsonsdisease .
Iron .Magnetic resonance imaging
Introduction
Iron accumulates in the brain in healthy aging; however, its
accumulation in specific regions is observed in neurodegener-
ative diseases including Parkinsons disease (PD) [1]. Specific
areas of the brain, such as globus pallidus (GB), substantia
nigra (SN), dentate nucleus (DN), and motor cortex (MC),
show increased mineralization in healthy brain. Neurons store
iron in the form of ferritin, which exists as light (L) and heavy
(H) chains. The iron redox couple mediates the transfer of
single electrons through the reversible oxidation/reduction re-
actions of Fe(II) and Fe(III). The biological redox potential,
electronic spin state as well as the reactivity of iron, is deter-
mined by the nature of the ligand to which the species are
bound. Iron configuration dictates whether an iron-based bio-
molecule is responsible for reactions involving oxygen trans-
port and storage, electron transfer, or oxidation/reduction of
other molecules [2]. Reactions involving iron in the body are
predominately redox-based, hydrolytic, or involve polynucle-
ar complex formation [3]. This is especially important for the
brain, where some of the highest concentrations of iron in the
body are maintained [4].
It has been hypothesized by a number of studies that nigral
mineralization may be a surrogate biomarker of PD. Mutations
in proteins involved in neuronal iron homeostasis have been
linked with PD, including transferrin (Tf) [5], iron regulatory
protein2(IRP2)[6], ferritin (Ft) [7], and divalent metal trans-
porter 1 (DMT1) [8]. In experimental animal models, direct
injection of iron in SN causes dopaminergic neurodegeneration
[9]. In addition, feeding neonatal mice with iron can trigger
parkinsonism and nigral degeneration [10].
Neurodegenerative disorders characterized by brain iron ac-
cumulation, like aceruloplasminemia [11,12],
neuroferritinopathy [13,14], and neurodegeneration with brain
iron accumulation (NBIA) [15] present parkinsonian symptoms.
*Sara Pietracupa
sara.pietracupa@uniroma1.it
1
IRCSS Neuromed, Pozzilli, Italy
2
Centre for Neuroinflammation and Neurodegeneration, Department
of Medicine, Imperial College London, London, UK
Neurol Sci
DOI 10.1007/s10072-017-3099-y
Loss-of-function mutations of IRPs show similar iron ac-
cumulation observed in idiopathic PD. Conversely, modula-
tion of iron content shows beneficial effects on PD animal
models. PD toxin model, 1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine (MPTP) or 6-hydroxydopamine (6-
OHDA) cause SN iron accumulation in mice. These PD
models show reduction of dopaminergic degeneration in SN
and increased striatal dopamine turnover after administration
of iron chelators [1618]. Iron-mediated toxicity in these
models can also be ameliorated by genetic or pharmacologi-
cally restoring Ft [16] and Cp [19].
The reason why iron accumulates in PD could be explained
by a number of iron-related proteins that are altered in PD. Ft
levels have been found to be decreased in postmortem PD
brains [20]; moreover, loss of iron storage capacity leads to
increased free unbound iron being more available for oxida-
tive reactions. Iron accumulation in PD might be caused by
increased neuronal iron import. DMT1 has been shown to be
elevated in the SN of postmortem PD brains [21]. This could
promote iron import, but the levels of transferrin receptor 1
(TfR1), which is required for DMT1-mediated iron import
were found unchanged when corrected for neuronal loss
[2224]. On the other hand, increased iron deposition might
also be attributed to reduced iron export. Although ceruloplas-
min (Cp) levels have been shown to beunaltered in PD brains,
however its activity is reduced in SN, which could narrow iron
efflux [19].
Furthermore, tau protein can play a role in iron accumula-
tion in PD [25]. A selective reduction oftau found in SN ofPD
patients may also contribute to iron accumulation by
preventing APP-mediated iron export [26].
Theroleofneuromalanin(NM)inironchelationhas
been recently proposed, NM binds iron in two different
high and low affinity sites. Most of the iron is bound to
high affinity sites, but under iron overload conditions, the
high affinity sites of NM are saturated, and iron binds to
the low affinity sites [27,28]. In the latter sites, iron is
sequesteredinthereactiveformandcouldplayadelete-
rious effect by promoting redox reactions when NM is
released to extraneuronal matrix. Therefore, the binding
of iron could have a protective role under physiological
conditions, but a toxic gain of function may manifest
when iron overload is present in the brain [29].
Neurotoxic interactions between iron and dopamine have
been recently proposed to explain degeneration of the SN
in PD [30]. Moreover, specific T1-weighted magnetic res-
onance imaging (MRI) sequence has been demonstrated
to detect NM signal changes to accurately discriminate
PD patients from controls. Nevertheless, the relationship
between iron accumulation and NM using multimodal im-
aging techniques has not found clear results [31].
Recent developments in MRI make it now possible to ex-
amine brain iron content in PD patients. Imaging studies have
shown increased iron accumulation at early stages of PD even
before symptom onset [3234]. The SN iron accumulation in
PD patients, shown by MRI, correlates with disease severity
[35,36] and duration. In this review, we will focus on the MRI
methods used for iron detection in PD. The Medline database
on PubMed was searched for relevant papers (last accessed on
the March 2017) using the following queries: BBrain iron
deposition^and BParkinsons disease^or BParkinsonism.^
We only considered in this review MRI studies, excluding
metabolic studies.
MRI methods to study iron accumulation
T2-weighted spin echo (SE) and fluid attenuation inversion
recovery (FLAIR) are the standard MRI protocol in neuro-
degenerative diseases showing hyperintensity in areas of
white matter (WM) lesions [37].FLAIRisusedtomaxi-
mize the detectability of lesions attenuating the signal gen-
erated by cerebrospinal fluid (CSF) [38]. Furthermore, SE-
based sequences have recently been modified with sam-
pling perfection with application optimized contrasts
(SPACE) using different flip-angle evolution [39,40].
This technique is a T2-weighted three-dimensional turbo
SE sequence with long echo time (TE) that enables fast
high-resolution three-dimensional images [39,40]. T2*-
weighted-GRE is also useful for detecting microbleeds
and calcifications [38]. Deep gray matter nuclei are well
presented in T2- and T2*-weighted images; however,
methods specifically designed for detecting iron-induced
susceptibility differences can be further improved by se-
quences like susceptibility-weighted imaging (SWI).
SWI is a novel high-resolution MRI modality which
[4143] requires a different from traditional spin-density,
T1, or T2 imaging recalled echo pulse sequence to acquire
images. The use of long TE allows for phase development
between the excitation pulse and data acquisition that is
sufficient for the differentiation of susceptibility variations
[41]. Due to its sensitivity, this method exploits the sus-
ceptibility differences between tissues and uses the phase
image to detect these [4244]. The magnitude and phase
data are combined to produce an enhanced contrast mag-
nitude image that is exquisitely sensitive to venous blood,
hemorrhage, and iron deposition. Furthermore,
susceptibility-weighted-angiography (SWAN) is useful to
precisely delineate small blood vessels, microbleeds, iron,
and calcium deposits. SWAN is innately less affected by
the chemical shift compared to SWI with significantly
enhanced susceptibility information. Finally, quantitative
susceptibility mapping (QSM) provides a novel contrast
mechanism in MRI [45] converting the phase shifts to
localize magnetic susceptibility.
Neurol Sci
Brain iron deposition in PD
In 1986, Drayer et al. [46] assessed for the first time the differ-
ences in T2 relaxation times in basal ganglia and SN in six
patients diagnosed with multi system atrophy (MSA) and pro-
gressive sopranuclear palsy (PSP) and 14 healthy controls
(HC). Authors found decreased T2 relaxation times in the pu-
tamen (PUT), and less prominent decreased in the caudate nu-
cleus (CN) and lateral pars compacta of the SN in the MSA and
PSP population compared with HC. Conversely, other authors
[47], studied postmortem brains of five subjects: four dying of
non-neurological conditions and one subject with PD by means
of T2-weighted images, showing no differences in iron distri-
bution in the basal ganglia between both study groups. Antonini
et al. [48] evaluated T2 relaxation times in 30 PD patients and
33 HC using a 1.5 T MRI. When compared to HC, PD patients
showed decreased T2 values in CN, putamen (PUT), and SN;
however, no clinical correlations were found with the afore-
mentioned regional increased iron accumulations.
In the last decade, several studies have focused their inter-
estinevaluatingironoverloadinPDpatients(Table1).
Increased evidence using SWI techniques confirm that parkin-
sonian patients show increased iron accumulation in deep
braingraynucleiwhencomparedtoHC[4952,55,59,60].
Several works [57,62] also demonstrated increased phase
values in the SN of PD positively correlating with UPDRS
motor scores (UPDRS III) [49] and Hoehn and Yahr (H&Y)
stage [55]aswellasadifferentironaccumulationinclinical
laterality [53]. In particular, Zhang et al. [49] investigated a
population of 42 PD patients and 30 HC analyzing SN and
basal ganglia. PD patients showed an increased deposition in
SN, PUT, and red nucleus (RN) bilaterally when compared to
HC. In addition, SN phase radians correlated with UPDRS III
of the most affected side. On the other hand, Wu and col-
leagues [55] demonstrated elevated iron deposition in PUT,
CN, RN, GP, and SN of 54 PD patients (18 PD patients with
HY < 1.5 and 36 PS patients with HY > 1.5) compared to 40
HC. The phase values of the GB and the SN showed negative
correlations with the HY stage. According to other data [50],
the regional phase values of 25 PD patients can reveal the
differences between PD patients with symmetric and
lateralized PD, even though the study included only a small
number of PD patients and HC. By contrast, other authors [59,
] found increased nigral mineralization in PD patients when
compared to HC; however, no correlations with motor sever-
ity as assessed with UPDRS III and disease stage were found.
In the study by Pechkam et al. [61], the authors addressed the
lack of significant correlations to the small number of PD
patients studied (18 patients). Previous results also suggest a
positive correlation between phase shifts values in SN and
serum ceruloplasmin levels in PD patients [52]in45PDpa-
tients (25 with decreased ceruloplasmin levels and 20 with
normal serum ceruplasmin levels and 45 HC). The original
finding in this study that PD patients with reduced ceruloplas-
min levels showed decreased nigral phase values when com-
pared with HC whilst PD patients with normal ceruloplasmin
levels demonstrated no changes in phase values when com-
pared with HC, suggests a role in iron deposition mediated by
ceruloplasmin levels. In a recent cross-sectional study [62], of
70 non-demented PD using high-pass filtered phase imaging,
PD patients showed higher levels of iron in SN, GP, and PUT
in PD patients compared to HC. Further correlations with
nigral mineralization were found with motor severity as
assessed with UPDRS III in off-medicated state and
bradykinesia-rigidity subscores. Moreover, when PD groups
were subdivided according to UPDRS-III score, iron mineral-
ization appears to be stratified according to disease motor
severity, confirming the idea that patients with a more severe
disease show a greater iron accumulation.
Other authors, Schwarz et al. (2014) [57], investigated the
usefulness of nigrosome-1 detection using SWI. Their sample
was divided as follows: a prospective case control study in 19
subjects (10 PD, 9 HC) and a retrospective cross-sectional
study in 95 consecutive patients (9 PD, 81 non-PD, 5 non-
Tabl e 1 Overview of studies using T2, T2*, SWI, and QSM sequence
in Parkinsons disease and other parkinsonisms
MR sequence HC PD
Drayer et al., [46] T2 GE 14 6 MSA
6PSP
Brooks et al., [47]T2 61PD
Antonini, [48]T2GE3330
Zhang et al. [49]T2GE
SWI
30 42
Grabner et al. [50]T1
SWI
525
Zhang et al. [51]T1
SWI
26 40
Jin et al. [52]T1
SWI
45 45
Wang et al. [53]T1IR
T1 FSE
T2 FLAIR
T2*ESWAN
14 20
Ulla et al. [54]T1
GRE
26 27
Wu et al. [55]T1
T2
SWI
54 40
Barbosa et al. [56]T1
R2 and R2 *
30 20
Schwarz et al. [57] SWI 90 19
He et al. [58] QSM 35 44
Guan et al. [59] QSM 40 60
Hopes et al. [60]T1
T2*
20 70
Peckham et al. [61] SWI 16 18
Martin Bastida et al. [62] SWI 20 70
Neurol Sci
diagnostic studies exclude). Two independent raters classified
subjects into PD and non-PD assessing the presence or ab-
sence of nigrosome-1. Almost 90% of participants were cor-
rectly classified, demonstrating a high sensitivity of the cur-
rent imaging sequence.
ESWAN sequences are also useful in detecting iron accu-
mulation. Wang et al. (2013) [53] demonstrated the presence
of smaller main MPV in the SN of PD patients as well as
negative correlation between the extent of reduction and se-
verity of motor symptoms, in a population of 20 PD patients
with different disease stages and 14 HC.
Recent works have been focused on evaluating the util-
ity of quantitative magnetic resonance (QMR) [56,58 and
59]. In a recent study (60) including 60 PD patients and 30
HC and comparing R2, R2* with quantitative susceptibil-
ity mapping (QSM), authors found QSM more accurate in
detecting iron accumulation. Further increased iron accu-
mulation measured with QSM in SN of PD patients but
also a spread in other structures such as RN and GP in
patients with a more advanced disease were found [59].
In addition, authors demonstrated positive correlation be-
tween QSM values and disease duration and UPDRS III in
a large number of subjects including 60 PD patients (45
withHY>2.5and15withHY>3)and40HC[58].
Finally, two longitudinal studies evaluated so far brain
iron overload in PD patients by means of R2* over a period
of 3 and 2 years, respectively [54,60]. In the first longitu-
dinal study [54], authors evaluated 26 PD patients and 26
HC by a first MRI whereas 14 PD patients and 18 HC
3 years after by a second MRI, whilst the second longitu-
dinal study included 70 PD patients in the cross-sectional
arm and 35 PD patients and 20 HC in the follow-up [60]. In
both studies, authors found significant changes in nigral
R2*valuesincomparisonwith HC which correlated with
changes in disease severity in the SN.
Discussion
In the last decade, a great number of studies evaluated iron
overload in parkinsonian disorders and other neurodegenera-
tive diseases. So far, there is accordance between authors that
iron is a promising biomarker for diagnosis in PD and other
parkinsonisms. However, the present studies present some
limitations. First of all, most of them are retrospective studies,
to our knowledge, only two longitudinal studies confirmed so
far, that iron overload increases as the disease progresses [54,
60]. Secondly, there is still lack of agreement between authors
on which is the most accurate sequence to study iron deposi-
tion. Indeed, a recent meta-analysis which compared postmor-
tem results with iron deposition detected by using MRI [63]
demonstrated that both R2* and SWI have some limitations in
evaluating iron deposition. This meta-analysis confirmed an
increased iron accumulation in SN and PUT as assessed by
R2*, SWI, and postmortem studies. On the other hand, whilst
both SWI and R2* indicated an increase in iron levels in the
RN, postmortem samples failed to find a correlation between
disease severity and iron deposition in RN. A number of con-
founding factors can influence imaging results. Calcifications,
microbleeds, and myelinated fibers can disrupt the iron signals
in both R* and SWI. These difficulties may be overcome
applying QSM sequences as previous studies already con-
firmed [53,56,57].
NM loss may be another confounding factor able to
reduce iron signals since in normal conditions it stores
big quantity of iron in the brain. A recent study [31]inves-
tigated the relationship between iron content in the SN and
neuromelanin signal changes combining T1 neuromelanin-
sensitive MRI sequence and T2* relaxometry. In this study,
iron content in the SN of PD patients did not show any
significant correlation with neuromelanin MRI signal
changes, which may indicate that neuromelanin reduction
and iron accumulation derive from different pathogenetic
mechanisms. MRI techniques sensitive to both NM chang-
es and iron accumulation have been hypothesize to be po-
tential surrogate biomarkers of PD [54,60,62,6469]Ina
recent study [69], 43 PD patients (13 late stage PD, 12 de
novo PD patients, 10 PD patients with a 25-year disease
duration, and 10 HS) were studied with NM MRI-sensi-
tive. Authors were able to identify a significant NM deple-
tion parallel with disease progression between PD patients
in an early stage of disease and groups of PD patients
belonging to more advanced disease stages. Moreover, oth-
er authors already found significant correlations between
NM loss and disease stage and severity [66]. On the other
hand, previous studies confirmed the role of iron as a
promising diagnostic biomarker in PD, finding an in-
creased iron accumulation in the SN correlating with dis-
ease duration and motor impairment [62]aswellasareli-
able marker of disease progression [54,60].
The use of iron sensitive MRI sequences may also be of
therapeutic interest. Iron chelators, such as deferiprone,
showed a disease-modifying effect in animal models of
PD [70]. So far, two randomized clinical trial with this
drug have been conducted in PD patients, showing mild
reductions in motor severity after 612 months of treat-
ment associated with iron removal in SN, CN, and DN
[71]. In the future, action of iron chelation and other drugs
can be followed with MRI as well as iron overload over
disease progression.
Conclusions
A number of studies have demonstrated that iron plays a role
in PD. Moreover, topographic distribution of iron could help
Neurol Sci
in some cases of parkinsonism in early stage differential diag-
nosis, even tough it is not clear if the increase in iron content in
PD is a pathogenetic or epiphenomenic event related to dopa-
minergic neurodegeneration. So far a limitation of previous
studies is that almost all of them have included only a rela-
tively small number of subjects. Larger studies are necessary
to confirm that iron can be considered as a novel biomarker in
PD and a new therapeutic target.
Compliance with ethical standards
Conflict of interest The authors declare that they have no conflict of
interest.
References
1. Thomas M, Jankovich J (2004) Neurodegenerative disease and iron
storage in the brain. Curr Opin Neurol 17(4):437442
2. Beard JL (2001) Iron biology in immune function, muscle metab-
olism and neuronal functioning. J Nutr 131(2S-2):568S579S
3. Aisen P, Enns C, Wessling-Resnick M (2001) Chemistry and biol-
ogy of eukaryotic iron metabolism. Int J Biochem Cell Biol 33(10):
940959
4. Gerlach M, Ben-Shachar D, Riederer P, Youdim MB (1994)
Altereted brain metabolism of iron as a cause of neurodegenerative
diseases? J Neurochem 63(3):793807
5. Borie C, Gasparini F, Verpillat P, Bonnet AM, Agid Y, Hetet G,
Brice A, Dürr A, Grandchamp B, French Parkinsons disease ge-
netic study group (2002) Association study between iron-related
genes polymorphisms and Parkinsons disease. J Neurol 249(7):
801804
6. Deplazes J, Schöbel K, Hochstrasser H, Bauer P, Walter U, Behnke
S, Spiegel J, Becker G, Riess O, Berg D (2004) Screening for
mutations of the IRP2 gene in Parkinsons disease patients with
hyperechogenicity of the substantia nigra. J Neural Transm
(Vienna) 111(4):515521
7. Foglieni B, Ferrari F, Goldwurm S, Santambrogio P, Castiglioni E,
Sessa M, Volontè MA, Lalli S, Galli C, Wang XS, Connor J, Sironi
F, Canesi M, Biasiotto G, Pezzoli G, Levi S, Ferrari M, Arosio P,
Cremonesi L (2007) Analysis offerritin genes in Parkinson disease.
Clin Chem Lab Med 45(11):14501456
8. He Q, Du T, Yu X, Xie A, Song N, Kang Q, Yu J, Tan L, Xie J,
Jiang H (2011) DMT1 polymorphism and risk of Parkinsonsdis-
ease. Neurosci Lett 501(3):128131. https://doi.org/10.1016/j.
neulet.2011.07.001
9. Ben-Shachar D, Youdim MB (1991) Intranigral iron injection in-
duces behavioral and biochemical Bparkinsonism^in rats. J
Neurochem 57(6):21332135
10. Kaur D, Peng J, Chinta SJ, Rajagopalan S, Di Monte DA, Cherny
RA, Andersen JK (2007) Increased murine neonatal iron intake
results in Parkinson-like neurodegeneration with age. Neurobiol
Aging 28(6):907913
11. Miyajima H, Nishimura Y, Mizoguchi K, Sakamoto M, Shimizu T,
Honda N (1987) Familial apoceruplasmin deficiency associated
with blepharospasm and retinal degeneration. Neurology 37(5):
761767
12. McNeill A, Pandolfo M, Kuhn J, Shang H, Miyajima H (2008) The
neurological presentation of ceruloplasmin gene mutations. Eur
Neurol 60(4):200205. https://doi.org/10.1159/000148691
13. Crompton DE, Chinnery PF, Fey C, Curtis AR, Morris CM,
Kierstan J, Burt A, Young F, Coulthard A, Curtis A, Ince PG,
Bates D, Jackson MJ, Burn J (2002) Neuroferrinopathy: a window
on the role of iron in neurodegeneration. Blood Cells Mol Dis
29(3):522531
14. Chinnery PF, Crompton DE, Birchall D, Jackson MJ, Coulthard A,
Lombès A, Quinn N, Wills A, Fletcher N, Mottershead JP, Cooper
P, Kellett M, Bates D, Burn J (2007) Clinical features and natural
history of neuroferritinopathy caused by the FTL1 460InsA muta-
tion. Brain 130(Pt 1):110119
15. Schneider SA, Hardy J, Bhatia KP (2012) Syndromes of neurode-
generationwith brain iron accumulation (NBIA): an update on clin-
ical presentations, histological and genetic underpinnings, and treat-
ment considerations. Mov Disord 27(1):4253. https://doi.org/10.
1002/mds.23971
16. Kaur D, Yantiri F, Rajagopalan S, Kumar J, Mo JQ, Boonplueang
R, Viswanath V, Jacobs R, Yang L, Beal MF, DiMonte D, Volitaskis
I, Ellerby L, Cherny RA, Bush AI, Andersen JK (2003) Genetic or
pharmacological iron chelation prevents MPTP-induced neurotox-
icity in vivo: a novel therapy for Parkinsons disease. Neuron 37(6):
899909
17. Mandel S, Maor G, Youdim MB (2004) Iron and alpha-synuclein in
the substantia nigra of MPTP-treated mice: effect of neuroprotec-
tive drugs R-apomorphine and green tea polyphenol ()-epigallo-
catechin-3-gallate. J Mol Neurosci 24(3):401416
18. Youdim MB, Stephenson G, Ben Shachar D (2004) Ironing iron out
in Parkinsons disease and other neurodegenerative diseases with
iron chelators: a lesson from 6-hydroxydopamine and iron chela-
tors, desferal and VK-28. Ann N YAcad Sci 1012:306325
19. Ayton S, Lei P, Bush AI (2013) Free Radic Biol Med 62:7689.
https://doi.org/10.1016/j.freeradbiomed.2012.10.558
20. Dexter DT, Carayon A, Vidailhet M, Ruberg M, Agid F, Agid Y,
Lees AJ, Wells FR, Jenner P, Marsden CD (1990) Decreased ferritin
levels in brain in Parkinsons disease. J Neurochem 55(1):1620
21. Salazar J, Mena N, Hunot S, Prigent A, Alvarez-Fischer D,
Arredondo M, Duyckaerts C, Sazdovitch V, Zhao L, Garrick LM,
Nuñez MT, Garrick MD, Raisman-Vozari R, Hirsch EC (2008)
Divalent metal transporter 1 (DMT1) contributes to neurodegener-
ation in animal models of Parkinsons disease. Proc Natl Acad Sci
U S A 105(47):1857818583. https://doi.org/10.1073/pnas.
0804373105
22. Mash DC, Pablo J, Buck BE, Sanchez-Ramos J, Weiner WJ (1991)
Distribution and number of transferrin receptors in Parkinsonsdis-
ease and in MPTP-treated mice. Exp Neurol 114(1):7381
23. Morris CM, Edwardson JA (1994) Iron histochemistry of the
substantia nigra in Parkinsons disease. Neurodegeneration 3(4):
277282
24. Faucheux BA, Hauw JJ, Agid Y, Hirsch EC (1997) The density of
[125I]-transferrin binding sites on perikarya of melanized neurons
of the substantia nigra is decreased in Parkinsons disease. Brain
Res 749(1):170174
25. Lei P, Ayton S, Finkelstein DI, Adlard PA, Masters CL, Bush AI
(2010) Tau protein: relevance to Parkinsons disease. Int J Biochem
Cell Biol 42(11):17751778. https://doi.org/10.1016/j.biocel.2010.
07.016
26. Lei P, Ayton S, Finkelstein DI, Spoerri L, Ciccotosto GD, Wright
DK, Wong BX, Adlard PA, Cherny RA, Lam LQ, Roberts BR,
Volitakis I, Egan GF, McLean CA, Cappai R, Duce JA, Bush AI
(2012) Tau deficiency induces parkinsonism with dementia by
impairing APP-mediated iron export. Nat Med 18(2):291295.
https://doi.org/10.1038/nm.2613
27. Zecca L, Casella L, Albertini A, Bellei C, Zucca FA, Engelen M,
Zadlo A, Szewczyk G, Zareba M, Sarna T (2008) Neuromelanin
can protect against iron-mediated oxidative damage in system
modeling iron overload of brain aging and Parkinsonsdisease.J
Neurochem 106(4):18661875. https://doi.org/10.1111/j.1471-
4159.2008.05541.x
Neurol Sci
28. Fasano M, Bergamasco B, Lopiano L (2006) Modifications of the
iron-neuromelanin system in Parkinsons disease. J Neurochem
96(4):909916
29. Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR (2004)
Iron, brain ageing and neurodegenerative disorders. Nat Rev
Neurosci 5(11):863873
30. Hare DJ, Double KL (2016) Iron and dopamine: a toxin couple.
Brain 139(Pt 4):10261035
31. Reimão S, Ferreira S, Nunes RG, Pita Lobo P, Neutel D, Abreu D,
Gonçalves N, Campos J, Ferreira JJ (2016) Magnetic resonance
correlation of iron content with neuromelanin inthe substantia nigra
of early stage Parkinsons disease. Eur J Neurol 23(2):368374.
https://doi.org/10.1111/ene.12838
32. Bartzokis G, Cummings JL, Markham CH, Marmarelis PZ,
Treciokas LJ, Tishler TA, Marder SR, Mintz J (1999) MRI evalu-
ation of brain iron in earlier- and later-onset Parkinsons diseaseand
normal subjects. Magn Reson Imaging 17(2):213222
33. Martin WR, Wieler M, Gee M (2008) Midbrain iron content in early
Parkinson disease: a potential biomarker of disease status.
Neurology 70(16 Pt 2):14111417. https://doi.org/10.1212/01.
wnl.0000286384.31050.b5
34. Berg D (2011) Substantia nigra hyperechogenicity is a risk marker
of Parkinsons disease: yes. J Neural Transm (Vienna) 118(4):613
619. https://doi.org/10.1007/s00702-010-0565-6
35. Wallis LI, Paley MN, Graham JM, Grünewald RA, Wignall EL, Joy
HM, Griffiths PD (2008) MRI assessment of basal ganglia iron
deposition in Parkinsons disease. J Magn Reson Imaging 28(5):
10611067. https://doi.org/10.1002/jmri.21563
36. Kosta P, Argyropoulou MI, Markoula S, Konitsiotis S (2006) MRI
evaluation of the basal ganglia size and iron content in patients with
Parkinson's disease. J Neurol 253(1):26
37. Bastos Leite AJ, van Straaten EC, Scheltens P, Lycklama G,
Barkhof F (2004) Thalamic lesions in vascular dementia: low sen-
sitivity of fluid-attenuated inversion recovery (FLAIR) imaging.
Stroke 35(2):415419
38. Hajnal JV, Bryant DJ, Kasuboski L, Pattany PM, De Coene B, Lewis
PD, Pennock JM, Oatridge A, Young IR, Bydder GM (1992) Use of
fluid attenuated inversion recovery (FLAIR) pulse sequences in MRI
of the brain. J Comput Assist Tomogr 16(6):841844
39. Mugler JP 3rd, Bao S, Mulkern RV, Guttmann CR, Robertson RL,
Jolesz FA, Brookeman JR (2000) Optimized single-slab three-di-
mensional spin-echo MR imaging of the brain. Radiology 216(3):
891899
40. Lichy MP, Wietek BM, Mugler JP 3rd, Horger W, Menzel MI,
Anastasiadis A, Siegmann K, Niemeyer T, Königsrainer A, Kiefer
B, Schick F, Claussen CD, Schlemmer HP (2005) Magnetic reso-
nance imaging of the body trunk using a single-slab, 3-dimensional,
T2-weighted turbo-spin-echo sequence with high sampling effi-
ciency (SPACE) for high spatial resolution imaging: initial clinical
experience. Investig Radiol 40(12):754760
41. Haacke EM, Mittal S, Wu Z, Neelavalli J, Cheng YC (2009)
Susceptibility-weighted imaging: technical aspects and clinical ap-
plications, part 1. AJNR Am J Neuroradiol30(1):1930. https://doi.
org/10.3174/ajnr.A1400
42. Haacke EM, Xu Y, Cheng YC, Reichenbach JR (2004)
Susceptibility weighted imaging (SWI). Magn Reson Med 52(3):
612618
43. Mittal S, Wu Z, Neelavalli J, Haacke EM (2009) Susceptibility-
weighted imaging: technical aspects and clinical applications, part
2. AJNR Am J Neuroradiol 30(2):232252. https://doi.org/10.
3174/ajnr.A1461
44. Mori N, Miki Y, Kasahara S, Maeda C, Kanagaki M, Urayama S,
Sawamoto N, Fukuyama H, Togashi K (2009) Susceptibility-
weighted imaging at 3 Tesla delineates the optic radiation.
Investig Radiol 44(3):140145.https://doi.org/10.1097/RLI.
0b013e318193ff25
45. Lotfipour AK, Wharton S, Schwarz ST, Gontu V, Schäfer A, Peters
AM, Bowtell RW, Auer DP, Gowland PA, Bajaj NP (2012) High
resolution magnetic susceptibility mapping of the substantia nigra
in Parkinsons disease. J Magn Reson Imaging 35(1):4855. https://
doi.org/10.1002/jmri.22752
46. Drayer BP, Olanow W, Burger P, Johnson GA, Herfkens R,
Riederer S (1986) Parkinson plus syndrome: diagnosis using high
field MR imaging of brain iron. Radiology 159(2):493498
47. Brooks DJ, Luthert P, Gadian D, Marsden CD (1989) Does signal-
attenuation on high-field T2-weighted MRI of the brain reflect re-
gional cerebral iron deposition? Observations on the relationship
between regional cerebral water proton T2 values and iron levels.
J Neurol Neurosurg Psychiatry 52(1):108111
48. Antonini A, Leenders KL, Meier D, Oertel WH, Boesiger P,
Anliker M (1993) T2 relaxation time in patients with Parkinsons
disease. Neurology 43(4):697700
49. Zhang W, Sun SG, Jiang YH, Qiao X, Sun X, Wu Y (2009)
Determination of brain iron content in patients with Parkinsons
disease using magnetic susceptibility imaging. Neurosci Bull
25(6):353360. https://doi.org/10.1007/s12264-009-0225-8
50. Grabner G, Haubenberger D, Rath J, Beisteiner R, Auff E, Trattnig
S, Barth M (2010) A population-specific symmetric phase modelto
automatically analyze susceptibility-weighted imaging (SWI) phase
shifts and phase symmetry in the human brain. J Magn Reson
Imaging 31(1):215220. https://doi.org/10.1002/jmri.22013
51. Zhang J, Zhang Y, Wang J, Cai P, Luo C, Qian Z, Dai Y, Feng H
(2010) Characterizing iron deposition in Parkinsons disease using
susceptibility-weighted imaging: an in vivo MR study. Brain Res
1330:124130. https://doi.org/10.1016/j.brainres.2010.03.036
52. Jin L, Wang J, Zhao L, Jin H, Fei G, Zhang Y, Zeng M, Zhong C
(2011) Decreased serum ceruloplasmin levels characteristically ag-
gravate nigral iron deposition in Parkinsons disease. Brain 134(Pt
1):5058. https://doi.org/10.1093/brain/awq319
53. Wang C, Fan G, Xu K, Wang S (2013) Quantitative assessment of
iron deposition in the midbrain using 3D-enhanced T2 star weight-
ed angiography (ESWAN): a preliminary cross-sectional study of
20 Parkinsons disease patients. Magn Reson Imaging 31(7):1068
1073. https://doi.org/10.1016/j.mri.2013.04.015
54. Ulla M, Bonny JM, Ouchchane L, Rieu I, Claise B, Durif F (2013)
Is R2* a new MRI biomarker for the progression of Parkinsons
disease? A longitudinal follow-up. PLoS One 8(3):e57904. https://
doi.org/10.1371/journal.pone.0057904
55. Wu SF, Zhu ZF, Kong Y, Zhang HP, Zhou GQ, Jiang QT, Meng XP
(2014) Assessment of cerebral iron content in patients with
Parkinsons disease by the susceptibility-weighted MRI. Eur Rev
Med Pharmacol Sci 18(18):26052608
56. Barbosa JH, Santos AC, Tumas V, Liu M, Zheng W, Haacke EM,
Salmon CE (2015) Quantifying brain iron deposition in patients
with Parkinsons disease using quantitative susceptibility mapping,
R2 and R2. Magn Reson Imaging 33(5):559565. https://doi.org/
10.1016/j.mri.2015.02.021
57. Schwarz ST, Afzal M, Morgan PS, Bajaj N, Gowland PA, Auer DP
(2014) The swallow tailappearance of the healthy nigrosomea
new accurate test of Parkinsons disease: a case-control and retro-
spective cross-sectional MRI study at 3T. PLoS One 9(4):e93814.
https://doi.org/10.1371/journal.pone.0093814
58. He N, Ling H, Ding B, Huang J, Zhang Y, Zhang Z, Liu C, Chen K,
Yan F (2015) Region-specific disturbed iron distribution in early
idiopathic Parkinsons disease measured by quantitative suscepti-
bility mapping. Hum Brain Mapp 36(11):44074420. https://doi.
org/10.1002/hbm.22928
59. GuanX,XuanM,GuQ,HuangP,LiuC,WangN,XuX,LuoW,
Zhang M (2016) Regionally progressive accumulation of iron in
Parkinsons diseaseas measured by quantitativesusceptibility map-
ping. NMR Biomed. https://doi.org/10.1002/nbm.3489
Neurol Sci
60. Hopes L, Grolez G, Moreau C, Lopes R, Ryckewaert G, Carrière N,
Auger F, Laloux C, Petrault M, Devedjian JC, Bordet R, Defebvre
L, Jissendi P, Delmaire C, Devos D (2016) Magnetic resonance
imaging features of the nigrostriatal system: biomarker of
Parkinsons disease stages? PLoS One 11(4):e0147947. https://
doi.org/10.1371/journal.pone.0147947
61. Peckham ME, Dashtipour K, Holshouser BA, Kani C, Boscanin A,
Kani K, Harder SL (2016) Novel pattern of iron deposition in the
fascicula nigrale in patients with Parkinsonsdisease.APilotStudy
2016:9305018. https://doi.org/10.1155/2016/9305018
62. Martin-Bastida A, Lao-Kaim NP, Loane C, Politis M, Roussakis
AA, Valle-Guzman N, Kefalopoulou Z, Paul-Visse G, Widner H,
Xing Y, Schwarz ST, Auer DP, Foltynie T, Barker RA, Piccini P
(2017) Motor associations of iron accumulation in deep grey matter
nuclei in Parkinsons disease: a cross-sectional study of iron-related
magnetic resonance imaging susceptibility. Eur J Neurol 24(2):
357365. https://doi.org/10.1111/ene.13208
63. Wang JY, Zhuang QQ, Zhu LB, Zhu H, Li T, Li R, Chen SF, Huang
CP, Zhang X, Zhu JH (2016) Meta-analysis of brain iron levels of
Parkinsons disease patients determined by postmortem and MRI
measurements. Sci Rep 6:36669. https://doi.org/10.1038/
srep36669
64. Kashihara K, Shinya T, Higaki F (2011) Neuromelanin magnetic
resonance imaging of nigral volume loss in patients with
Parkinsons disease. J Clin Neurosci 18(8):10931096. https://doi.
org/10.1016/j.jocn.2010.08.043
65. Schwarz ST, Rittman T, Gontu V, Morgan PS, Bajaj N, Auer DP
(2011) T1-weighted MRI shows stage-dependent substantia nigra
signal loss in Parkinsons disease. Mov Disord 26(9):16331638.
https://doi.org/10.1002/mds.23722
66. Matsuura K, Maeda M, Yata K, Ichiba Y, Yamaguchi T, Kanamaru
K, Tomimoto H (2013) Neuromelanin magnetic resonance imaging
in Parkinsons disease and multiple system atrophy. Eur Neurol
70(12):7077. https://doi.org/10.1159/000350291
67. Miyoshi F, Ogawa T, Kitao SI, Kitayama M, Shinohara Y, Takasugi
M, Fujii S, Kaminou T (2013) Evaluation of Parkinson disease and
Alzheimer disease with the use of neuromelanin MR imaging and
(123)I-metaiodobenzylguanidine scintigraphy. AJNR Am J
Neuroradiol Nov-Dec 34(11):21132118. https://doi.org/10.3174/
ajnr.A3567
68. Castellanos G, Fernández-Seara MA, Lorenzo-Betancor O, Ortega-
Cubero S, Puigvert M, Uranga J, Vidorreta M, Irigoyen J, Lorenzo
E, Muñoz-Barrutia A, Ortiz-de-Solorzano C, Pastor P, Pastor MA
(2015) Automated neuromelanin imaging as a diagnostic biomarker
for Parkinsons disease. Mov Disord 30(7):945952. https://doi.
org/10.1002/mds.26201
69. Fabbri M, Reimão S, Carvalho M, Nunes RG, Abreu D, Guedes
LC, Bouça R, Lobo PP, Godinho C, Coelho M, Gonçalves NC,
Rosa MM, Antonini A, Ferreira JJ (2017) Substantia Nigra
Neuromelanin as an imaging biomarker of disease progression in
Parkinsons disease. J Parkinsons Dis. https://doi.org/10.3233/JPD-
171135
70. Dexter DT, Statton SA, Whitmore C, Freinbichler W, Weinberger P,
Tipton KF, Della Corte L, Ward RJ, Crichton RR (2011) Clinically
available iron chelators induce neuroprotection in the 6-OHDA
model of Parkinsons disease after peripheral administration. J
Neural Transm (Vienna) 118(2):223231. https://doi.org/10.1007/
s00702-010-0531-3
71. Devos D, Moreau C, Devedjian JC, Kluza J, Petrault M, Laloux C,
Jonneaux A, Ryckewaert G, Garçon G, Rouaix N, Duhamel A,
Jissendi P, Dujardin K, Auger F, Ravasi L, Hopes L, Grolez G,
Firdaus W, Sablonnière B, Strubi-Vuillaume I, Zahr N, Destée A,
Corvol JC, Pöltl D, Leist M, Rose C, Defebvre L, Marchetti P,
Cabantchik ZI, Bordet R (2014) Targeting chetable iron as a thera-
peutic modality in Parkinsons disease. Antioxid Redox Signal
21(2):195210. https://doi.org/10.1089/ars.2013.5593
Neurol Sci
... The interaction between water molecules and paramagnetic iron complexes, such as ferritin and hemosiderin, contribute to the transversal magnetization dephasing [11]. Being so, T2-weighted spin-echo and gradient echo (GRE), as well as T2 * -weighted GRE MRI techniques, have been extensively used in the study of deep gray matter nuclei in order to evaluate the presence of iron accumulation [12]. Consequently, the paramagnetic iron causes a progressive signal reduction in T2 and T2 * -weighted MRI, proportionally to its concentration [13] (Figure 1). ...
... While in the same study, SWI hypointensity in the putamen was significantly correlated with the obtained Tinetti total score [88]. 12 Behavioural Neurology A significant increase in SN susceptibility on 3.0 T enhanced T2 * -weighted angiography scanning (ESWAN) has been reported in PD patients, both in tremor dominant and akinetic/rigid variants [89]. In fact, the ESWAN is a 3D multi-echo gradient-echo pulse sequence with partial flow compensation, using multiple magnitude or phase images with different echo times for image generation: the first echo applies the arterial inflow effect, whereas longer echoes are responsible for susceptibility effects. ...
Article
Full-text available
Brain iron load is one of the most important neuropathological hallmarks in movement disorders. Specifically, the iron provides most of the paramagnetic metal signals in the brain and its accumulation seems to play a key role, although not completely explained, in the degeneration of the basal ganglia, as well as other brain structures. Moreover, iron distribution patterns have been implicated in depicting different movement disorders. This work reviewed current literature on Magnetic Resonance Imaging for Brain Iron Detection and Quantification (MRI-BIDQ) in neurodegenerative processes underlying movement disorders.
... Magnetic resonance imaging (MRI) is a reliable, noninvasive method for detecting brain iron deposition in patients with PD and does not harm the human body (14). A number of magnetic resonance (MR) methods have been used to measure the iron levels in the brain, including susceptibility-weighted imaging (SWI); R2, R2*, and R2' phase; and quantitative susceptibility mapping (QSM) (15). ...
Article
Full-text available
Background Quantitative susceptibility mapping (QSM) is a novel imaging method for detecting iron content in the brain. The study aimed determine whether the iron deposition in the brains of people with Parkinson’s disease (PD) is correlated with freezing of gait (FOG). Methods We retrospectively collected the data of 24 patients with PD from the Movement Disorders Program and 36 healthy controls (HCs) from January 2021 to December 2021. Clinical assessments included mental intelligence scales, Parkinson rating scales, motor-related scales, and clinical gait assessments. All exercise scales and gait assessments were performed in the “ON” and “OFF” states. Magnetic resonance imaging (MRI) data were collected using 3-dimensional fast low-angle shot sequences. We chose the bilateral red nucleus, substantia nigra, thalamus, putamen, caudate nucleus, and globus pallidus as regions of interest for QSM analysis. Results The iron deposition in the substantia nigra of the PD group was significantly higher than that of the HC group (P<0.01). In the PD group, the iron deposition in the substantia nigra of patients with FOG was significantly higher than that in patients without FOG (P=0.04). The iron deposition in the substantia nigra was positively correlated with the New Freezing of Gait Questionnaire (P=0.03). The scores for depression and anxiety of the PD group were significantly higher than those of the HC group, while the Berg balance scale score was significantly lower (P<0.01). Conclusions The iron deposition in the substantia nigra of patients with PD is increased compared with that of controls and is associated with FOG. QSM can be used to detect brain iron deposition in patients with PD, which would help to explore the mechanism of abnormal neurobiological activity in FOG.
... However, "in vivo" MRI observations have to be validated by post-mortem examination [7] . T2 and T2*-weighted MRI images are currently the preferred methods to visualize and quantify the amount of Fe in the deep brain structures [8,9] . The present post-mortem 7.0-tesla MRI study investigates and semi-quantifies the amount of Fe in different structures of the brains of patients with Parkinson's disease and atypical parkinsonian syndromes compared to normal controls. ...
... Parkinson's disease (PD) is a devastating neurological condition marked by stiffening, tremors, and difficulty with walking, and even a variety of non-motor manifestations. The preferential death of dopaminergic neuronal cells in the substantia nigra pars compacta (SNpc) is a critical aspect in this neurological illness (Berg & Youdim, 2006;Muñoz et al., 2016; and the elevated iron levels inside the affected area mainly SNpc, which houses deteriorating dopaminergic neuronal cells, are a well-known characteristic of Parkinson's disease (Sofic et al., 1988;Andersen, 2004;Pietracupa et al., 2017;Cheng et al., 2020a, b). In PD, the quantity of proteins, which regulates iron concentration, is abnormally shifted, leading to greater labile iron pool, which also can cause synuclein agglomeration [1][2][3][4][5][6][7][8][9][10][11][12][13][14][15]91]. ...
... Parkinson's disease (PD) is a devastating neurological condition marked by stiffening, tremors, and difficulty with walking, and even a variety of non-motor manifestations. The preferential death of dopaminergic neuronal cells in the substantia nigra pars compacta (SNpc) is a critical aspect in this neurological illness (Berg and Youdim 2006;Muñoz et al. 2016;) and the elevated iron levels inside the affected area mainly SNpc, which houses deteriorating dopaminergic neuronal cells, are a well-known characteristic of Parkinson's disease (Sofic et al. 1988;Andersen 2004;Pietracupa et al. 2017;Cheng et al. 2020). In PD, the quantity of proteins, which regulates iron concentration, is abnormally shifted, leading to greater labile iron pool, which also can cause synuclein agglomeration [1][2][3][4][5][6][7][8][9][10][11][12][13][14][15]91]. ...
Chapter
Full-text available
Iron and calcium have one thing in common: they are both required for appropriate neural activity. Neuronal calcium impulses drive neurochemical release, proliferation of axon and synapse formation, and govern the gene expression involved in memory formation, while, iron is essential for normal functioning of mitochondria, synapse formation, and the acquisition of cognitive skills. Latest research has discovered that cellular iron enhances calcium signalling, resulting in subsequent stimulation of kinase pathways. Moreover, iron-mediated reactive oxygen species production strengthens usual calcium-regulated signalling processes under physiological environment, whereas, elevated iron levels induces oxidative stress, which leads to an increase in uncontrolled calcium inputs that potentially harm normal functioning of mitochondria and other associated functions. In this chapter, we will examine the facts surrounding a poorly known link between iron and calcium balance and connection, in which one’s deregulation has an affect on another, resulting in a negative feedback system that eventually leads to neurodegenerative disorders. We provides a brief account on background, homeostasis of iron and calcium and related anomalies upon imbalances occur. Also describe the crosstalk among these two i.e. iron and calcium and related cell death (ferroptosis) and their connecting links.
... At present, the pathogenesis of PD is not clear (5). Some studies have shown that the substantia nigra (SN) is one of the important structures in the pathophysiology of PD, and can even be considered as the only credible brain structure closely related to neuron loss (6). ...
Article
Full-text available
Parkinson's disease (PD) is a common nervous system disease, mainly manifested as motor retardation, resting tremor, etc. (1). The clinical features of early PD patients are not characteristic, and diagnosis is very difficult. When obvious PD manifestations are found, the number of dopaminergic neurons in substantia nigra of patients has been reduced by more than half, and the treatment is difficult (2). Early diagnosis or auxiliary diagnosis of PD in clinical work is crucial for the treatment of PD and the prognosis of patients. In recent years, cerebral ultrasound has been widely used in the diagnosis and treatment of some diseases, such as Parkinson's disease, Alzheimer's disease, tuberculous meningitis, brain injury, etc., especially for the study of PD. The European Union of neuroscience and the latest diagnostic guidelines for PD in China have confirmed the role of the transcranial sonography (TCS). This article reviews the recent advances in the study of PD by transcranial sonography.
... Iron-sensitive MRI techniques, such as relaxometry (T2* or R2*) and susceptibility-weighted imaging (SWI), revealed increased nigral iron accumulation in PD, which was associated with disease severity in most studies 50 . However, the sensitivity and specificity of the techniques were variable, and heterogeneous results were obtained in extra-nigral deep nuclei. ...
Article
Parkinson disease (PD) is a progressive disorder characterized by dopaminergic neurodegeneration in the brain. The development of parkinsonism is preceded by a long prodromal phase, and >50% of dopaminergic neurons can be lost from the substantia nigra by the time of the initial diagnosis. Therefore, validation of in vivo imaging biomarkers for early diagnosis and monitoring of disease progression is essential for future therapeutic developments. PET and single-photon emission CT targeting the presynaptic terminals of dopaminergic neurons can be used for early diagnosis by detecting axonal degeneration in the striatum. However, these techniques poorly differentiate atypical parkinsonian syndromes from PD, and their availability is limited in clinical settings. Advanced MRI in which pathological changes in the substantia nigra are visualized with diffusion, iron-sensitive susceptibility and neuromelanin-sensitive sequences potentially represents a more accessible imaging tool. Although these techniques can visualize the classic degenerative changes in PD, they might be insufficient for phenotyping or prognostication of heterogeneous aspects of PD resulting from extranigral pathologies. The retina is an emerging imaging target owing to its pathological involvement early in PD, which correlates with brain pathology. Retinal optical coherence tomography (OCT) is a non-invasive technique to visualize structural changes in the retina. Progressive parafoveal thinning and fovea avascular zone remodelling, as revealed by OCT, provide potential biomarkers for early diagnosis and prognostication in PD. As we discuss in this Review, multimodal imaging of the substantia nigra and retina is a promising tool to aid diagnosis and management of PD.
... Increased iron content in the substantia nigra pars compacta, where degenerating neuromelanin-bearing dopaminergic neurons reside, is a well-established feature of PD [87][88][89][90]. In agreement with the observations reported for AD, the content of iron-regulatory proteins is aberrantly altered in PD, resulting in a higher labile iron pool that can also provoke α-synuclein aggregation [15,91]. ...
Article
Iron, through its participation in oxidation/reduction processes, is essential for the physiological function of biological systems. In the brain, iron is involved in the development of normal cognitive functions, and its lack during development causes irreversible cognitive damage. Yet, deregulation of iron homeostasis provokes neuronal damage and death. Ferroptosis, a newly described iron-dependent cell death pathway, differs at the morphological, biochemical, and genetic levels from other cell death types. Ferroptosis is characterized by iron-mediated lipid peroxidation, depletion of the endogenous antioxidant glutathione and altered mitochondrial morphology. Although iron promotes the emergence of Ca²⁺ signals via activation of redox-sensitive Ca²⁺ channels, the role of Ca²⁺ signaling in ferroptosis has not been established. The early dysregulation of the cellular redox state observed in ferroptosis is likely to disturb Ca²⁺ homeostasis and signaling, facilitating ferroptotic neuronal death. This review presents an overview of the role of iron and ferroptosis in neuronal function, emphasizing the possible involvement of Ca²⁺ signaling in these processes. We propose, accordingly, that the iron-ferroptosis-Ca²⁺ association orchestrates the progression of cognitive dysfunctions and memory loss that occurs in neurodegenerative diseases. Therefore, to prevent iron dyshomeostasis and ferroptosis, we suggest the use of drugs that target the abnormal Ca²⁺ signaling caused by excessive iron levels as therapy for neurological disorders.
Article
Full-text available
The brain is the most complex organ in the human body and, as such, its study entails great challenges (methodological, theoretical, etc.). Nonetheless, there is a remarkable amount of studies about the consequences of pathological conditions on its development and functioning. This bibliographic review aims to cover mostly findings related to changes in the physical distribution of neurons and their connections—the connectome—both structural and functional, as well as their modelling approaches. It does not intend to offer an extensive description of all conditions affecting the brain; rather, it presents the most common ones. Thus, here, we highlight the need for accurate brain modelling that can subsequently be used to understand brain function and be applied to diagnose, track, and simulate treatments for the most prevalent pathologies affecting the brain.
Article
Full-text available
Background and purpose: To determine whether iron deposition in deep brain nuclei assessed using high-pass filtered phase imaging plays a role in motor disease severity in Parkinson's disease (PD). Methods: Seventy patients with mild to moderate PD and 20 age- and gender-matched healthy volunteers (HVs) underwent susceptibility-weighted imaging on a 3 T magnetic resonance imaging scanner. Phase shifts (radians) in deep brain nuclei were derived from high-pass filtered phase images and compared between groups. Analysis of clinical laterality and correlations with motor severity (Unified Parkinson's Disease Rating Scale, Part III, UPDRS-III) were performed. Phase shifts (in radians) were compared between HVs and three PD subgroups divided according to UPDRS-III scores using analysis of covariance, adjusting for age and regional area. Results: Parkinson's disease patients had significantly (P < 0.001) higher radians than HVs bilaterally in the putamen, globus pallidus and substantia nigra (SN). The SN contralateral to the most affected side showed higher radians (P < 0.001) compared to the less affected side. SN radians positively correlated with UPDRS-III and bradykinesia-rigidity subscores, but not with tremor subscores. ancova followed by post hoc Bonferroni-adjusted pairwise comparisons revealed that SN radians were significantly greater in the PD subgroup with higher UPDRS-III scores compared to both lowest UPDRS-III PD and HV groups (P < 0.001). Conclusions: Increased nigral iron accumulation in PD appears to be stratified according to disease motor severity and correlates with symptoms related to dopaminergic neurodegeneration. This semi-quantitative in vivo iron assessment could prove useful for objectively monitoring PD progression, especially in clinical trials concerning iron chelation therapies.
Article
Full-text available
Brain iron levels in patients of Parkinson’s disease (PD) are usually measured in postmortem samples or by MRI imaging including R2* and SWI. In this study we performed a meta-analysis to understand PD-associated iron changes in various brain regions, and to evaluate the accuracy of MRI detections comparing with postmortem results. Databases including Medline, Web of Science, CENTRAL and Embase were searched up to 19th November 2015. Ten brain regions were identified for analysis based on data extracted from thirty-three-articles. An increase in iron levels in substantia nigra of PD patients by postmortem, R2* or SWI measurements was observed. The postmortem and SWI measurements also suggested significant iron accumulation in putamen. Increased iron deposition was found in red nucleus as determined by both R2* and SWI, whereas no data were available in postmortem samples. Based on SWI, iron levels were increased significantly in the nucleus caudatus and globus pallidus. Of note, the analysis might be biased towards advanced disease and that the precise stage at which regions become involved could not be ascertained. Our analysis provides an overview of iron deposition in multiple brain regions of PD patients, and a comparison of outcomes from different methods detecting levels of iron.
Article
Full-text available
Background and Purpose. To determine whether the pattern of iron deposition in the fascicula nigrale in patients with Parkinson’s disease would be different from age-matched controls by utilizing quantitative susceptibility mapping to measure susceptibility change. Methods. MRIs of the brain were obtained from 34 subjects, 18 with Parkinson’s disease and 16 age- and gender-matched controls. Regions of interest were drawn around the fascicula nigrale and substantia nigra using SWI mapping software by blinded investigators. Statistical analyses were performed to determine susceptibility patterns of both of these regions. Results. Measurements showed significantly increased susceptibility in the substantia nigra in Parkinson’s patients and an increased rostral-caudal deposition of iron in the fascicula nigrale in all subjects. This trend was exaggerated with significant correlation noted with increasing age in the Parkinson group. Conclusion. The pattern of an exaggerated iron deposition gradient of the fascicula nigrale in the Parkinson group could represent underlying tract dysfunction. Significant correlation of increasing iron deposition with increasing age may be a cumulative effect, possibly related to disease duration.
Article
Full-text available
Introduction: Magnetic resonance imaging (MRI) can be used to identify biomarkers in Parkinson's disease (PD); R2* values reflect iron content related to high levels of oxidative stress, whereas volume and/or shape changes reflect neuronal death. We sought to assess iron overload in the nigrostriatal system and characterize its relationship with focal and overall atrophy of the striatum in the pivotal stages of PD. Methods: Twenty controls and 70 PD patients at different disease stages (untreated de novo patients, treated early-stage patients and advanced-stage patients with L-dopa-related motor complications) were included in the study. We determined the R2* values in the substantia nigra, putamen and caudate nucleus, together with striatal volume and shape analysis. We also measured R2* in an acute MPTP mouse model and in a longitudinal follow-up two years later in the early-stage PD patients. Results: The R2* values in the substantia nigra, putamen and caudate nucleus were significantly higher in de novo PD patients than in controls. Early-stage patients displayed significantly higher R2* values in the substantia nigra (with changes in striatal shape), relative to de novo patients. Measurements after a two-year follow-up in early-stage patients and characterization of the acute MPTP mouse model confirmed that R2* changed rapidly with disease progression. Advanced-stage patients displayed significant atrophy of striatum, relative to earlier disease stages. Conclusion: Each pivotal stage in PD appears to be characterized by putative nigrostriatal MRI biomarkers: iron overload at the de novo stage, striatal shape changes at early-stage disease and generalized striatal atrophy at advanced disease.
Article
Full-text available
View largeDownload slide Iron accumulation has long been associated with neurodegeneration in Parkinson’s disease. Double and Hare describe how iron and dopamine form a toxic redox couple that creates a hazardous chemical environment inside cells. This process may represent both an upstream mechanism of disease, and a viable target for new therapies. View largeDownload slide Iron accumulation has long been associated with neurodegeneration in Parkinson’s disease. Double and Hare describe how iron and dopamine form a toxic redox couple that creates a hazardous chemical environment inside cells. This process may represent both an upstream mechanism of disease, and a viable target for new therapies.
Article
Full-text available
The progression of Parkinson's disease (PD) seems to vary according to the disease stage, which greatly influences the management of PD patients. However, the underlying mechanism of progression in PD remains unclear. This study was designed to explore the progressive pattern of iron accumulation at different stages in PD patients. Sixty right-handed PD patients and 40 normal controls were recruited. According to the disease stage, 45 patients with Hoehn-Yahr stage ≤ 2.5 and 15 patients with Hoehn-Yahr stage ≥ 3 were grouped into early-stage PD (EPD) and late-stage PD (LPD) groups, respectively. The iron content in the cardinal subcortical nuclei covering the cerebrum, cerebellum and midbrain was measured using quantitative susceptibility mapping (QSM). The substantia nigra pars compacta (SNc) showed significantly increased QSM values in the EPD patients compared with the controls. In the LPD patients, while the SNc continued to show increased QSM values compared with the controls and EPD patients, the regions showing increased QSM values spread to include the substantia nigra pars reticulata (SNr), red nucleus (RN) and globus pallidus (GP). Our data also indicated that iron deposition was more significant in the GP internal segment (GPi) than in the GP external segment. No other regions showed significant changes in QSM values among the groups. Therefore, we were able to confirm a regionally progressive pattern of iron accumulation in the different stages of PD, indicating that iron deposition in the SNc is affected exclusively in the early stages of the disease, while the SNr, RN and GP, and particularly the GPi segment, become involved in advanced stages of the disease. This is a preliminary study providing objective evidence of the iron-related progression in PD. Copyright © 2016 John Wiley & Sons, Ltd.
Article
Background: A specific T1-weighted magnetic resonance imaging (MRI) sequence has been shown to detect substantia nigra (SN) neuromelanin (NM) signal changes that accurately discriminate Parkinson's disease (PD) patients from controls, even in early disease stages. However, it is unclear what happens to these SN changes in later disease stages and if they can be a marker of disease progression. Objective: to investigate the pattern of SN-NM area loss and contrast ratio (CR) intensity changes in late-stage PD (LSPD) compared to earlier disease stages. Methods: A comparative cross-sectional study was performed, analyzing SN-NM MRI signal in LSPD (Schwab and England Activities of Daily Living Scale score <50 or Hoehn Yahr Stage [HY] >3), comparing this group with de novo, 2-5 year PD and controls. SN-NM signal area and CR values for the internal and lateral SN regions were obtained with semi-automated methods. Results: 13 LSPD, 12 de novo patients with PD, 10 PD patients with a 2-5 year disease duration, and 10 controls were included. NM signal area was significantly decreased in LSPD compared to de novo PD (P-value = 0.005; sensitivity: 75%; specificity 92% and AUC: 0.86). In the lateral SN region, a decrease in the CR was detected in all PD groups compared to controls; despite not reaching statistical significance, a slight increment was observed comparing LSPD to 2-5 year PD. NM signal area significantly correlated with HY (R = -0.37; P < 0.05) and Movement disorder Society Unified Parkinson's Disease Rating Scale part II (MDS-UPDRS) (R = -0.4; P < 0.05) while a weak correlation was found with MDS-UPDRS part III (R = -0.26; P: 0.1). Conclusion: SN area evaluated by NM-sensitive MRI may be a promising biomarker of nigral degeneration and disease progression in PD patients.
Article
Background and purpose: Magnetic resonance (MR) studies have demonstrated a significant reduction of neuromelanin in the substantia nigra (SN) of Parkinson's disease (PD) patients with high accuracy for differential diagnosis compared to non-PD controls and essential tremor. However, studies state that not knowing how paramagnetic effects of iron influence neuromelanin signal is a limitation. In this study a neuromelanin-sensitive MR sequence was combined with T2* relaxometry iron quantification analysis to study the SN of early-stage PD patients to investigate the correlation between these parameters. Methods: The inclusion criteria were untreated de novo PD patients and a 2-5 year disease duration (early PD); in addition, age-matched controls were enrolled. These were studied at 3.0 T with a high-resolution T1-weighted MR sequence to visualize neuromelanin and a relaxometry sequence for iron quantification. The primary outcome was the correlation of the width of the neuromelanin high signal region and the T2* values in the lateral, central and medial segments of the SN. Results: Very weak correlations of T2* values with neuromelanin width, positive for global and negative for the medial and lateral SN segments, were found in both PD groups and control subjects. The SN neuromelanin width was markedly reduced in the de novo and early PD groups compared with controls in all SN segments, but no significant difference in T2* values was found between the groups. Conclusions: The SN neuromelanin signal does not have a significant correlation with iron content in PD patients or controls. The neuromelanin MR signal reduction in PD does not seem to be significantly influenced by paramagnetic iron effects.
Article
In Parkinson's disease (PD), iron elevation in specific brain regions as well as selective loss of dopaminergic neurons is a major pathologic feature. A reliable quantitative measure of iron deposition is a potential biomarker for PD and may contribute to the investigation of iron-mediated PD. The primary purpose of this study is to assess iron variations in multiple deep grey matter nuclei in early PD with a novel MRI technique, quantitative susceptibility mapping (QSM). The inter-group differences of susceptibility and R2(*) value in deep grey matter nuclei, namely head of caudate nucleus (CN), putamen (PUT), global pallidus (GP), substantia nigra (SN), and red nucleus (RN), and the correlations between regional iron deposition and the clinical features were explored in forty-four early PD patients and 35 gender and age-matched healthy controls. Susceptibility values were found to be elevated within bilateral SN and RN contralateral to the most affected limb in early PD compared with healthy controls (HCs). The finding of increased susceptibility in bilateral SN is consistent with work on a subgroup of patients at the earliest clinical detectable state (Hoehn and Yahr [1967]: Neurology 17:427-442; Stage I). However, increased R2(*) values were only seen within SN contralateral to the most affected limb in the PD group when compared with controls. Furthermore, bilateral SN magnetic susceptibility positively correlated with disease duration and UPDRS-III scores in early PD. This finding supports the potential value of QSM as a non-invasive quantitative biomarker of early PD. Hum Brain Mapp, 2015. © 2015 Wiley Periodicals, Inc. © 2015 Wiley Periodicals, Inc.