Skip to main content
Erschienen in: Brain Structure and Function 7/2022

Open Access 19.07.2022 | Review

Nigral neuropathology of Parkinson’s motor subtypes coincide with circuitopathies: a scoping review

verfasst von: Jackson Tyler Boonstra, Hugo McGurran, Yasin Temel, Ali Jahanshahi

Erschienen in: Brain Structure and Function | Ausgabe 7/2022

Abstract

The neuropathological substrates of Parkinson’s disease (PD) patients with motor subtypes tremor-dominance (TD), non-tremor dominance (nTD), postural instability and gait difficulty (PIGD), and akinetic-rigid (AR) are not completely differentiated. While extensive pathological research has been conducted on neuronal tissue of PD patients, data have not been discussed in the context of mechanistic circuitry theories differentiating motor subtypes. It is, therefore, expected that a more specific and tailored management of PD symptoms can be accomplished by understanding symptom-specific neuropathological mechanisms with the detail histology can provide. This scoping review gives an overview of the literature comparing TD and nTD PD motor subtypes by clarify observed pathology with underlying physiological circuitry theories. Studies using an array of pathological examination techniques have shown significant differences between TD and nTD PD subtypes. nTD PD patients show higher neuronal loss, gliosis, extraneuronal melanin deposits, and neuroaxonal dystrophy in multiple subregions of the substantia nigra (SN) related to the overactivity of the indirect motor loop. TD patients show more severe cell loss specifically in medial SN subdivisions, and have damage in the retrorubral field A-8 that projects to the dorsolateral striatum and ventromedial thalamus in the direct motor loop. Pathological studies are consistent with neuroimaging data and support contemporary mechanistic circuitry theories of PD motor symptom genesis. Further multimodal neuroimaging and histological studies are required to validate and expand upon these findings.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00429-022-02531-9.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Parkinson’s disease (PD) is a progressive neurodegenerative disorder that affects more than 10 million people worldwide (Marras et al. 2018). Clinical diagnoses of PD are based on the identification of cardinal motor symptoms including bradykinesia, postural instability, rigidity, and tremor. Motor symptom subtypes can be clinically determined with the Movement Disorder Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS), an assessment of motor and non-motor PD symptoms (Goetz et al. 2007). The ratio of the mean tremor score to the mean score of their postural instability and gait difficulty (PIGD) score can be used to differentiate tremor-dominance (TD) patients (ratio ≥ 1.5), from PIGD patients (ratio ≤ 1), and from intermediate or ‘mixed-type’ patients (ratios > 1.0 and < 1.5) (Jankovic et al. 1990). Those patients diagnosed with PIGD have been called akinetic-rigid (AR) with both subtypes often denoted as non-tremor dominant (nTD).
Heterogeneous motor symptom presentation between PD motor subtypes suggest correspondingly diverse pathophysiologies (He et al. 2017; Rajput et al. 2009; van Rooden et al. 2010; Zaidel et al. 2009). Previous research reviews both TD and nTD PD patients differing in various neuroanatomical and neurofunctional domains (Boonstra et al. 2021) as well as in non-motor symptoms and quality of life (Wu et al. 2016). Patients with nTD have shown accelerated motor, functional, and cognitive decline compared to TD, resulting in TD to be considered the less severe subtype (Wu et al. 2016). In line with differing symptoms and disease severity, histological data show TD and nTD subtypes have ‘specific morphological lesion patterns of pathophysiological relevance’ (Jellinger and Paulus 1992; Paulus and Jellinger 1991). It is, therefore, expected that a more specific and tailored management of PD symptoms can be accomplished by understanding symptom-specific neuropathological mechanisms with the detail histology can provide. Pathological examinations further contribute to the validation of in vivo neuroimaging readouts by virtue of resolution as the imaging modalities assist each other in a complementary nature.
Despite a plethora of research investigating the pathophysiological bases of PD (Borghammer 2018; Braak et al. 2003; Hornykiewicz 1998), particularly regarding the degeneration of midbrain dopamine (DA) neurons in the substantia nigra pars compacta (SNc), the full extent of PD pathology is poorly understood and even less is known about pathological differences between PD motor subtypes. Moreover, how previously described pathophysiological alterations between PD motor subtypes fit into contemporary mechanistic circuitry theories of PD symptoms genesis has yet to be described. The aim of this scoping review is to fill the gap between pathology and circuitry by consolidating previous studies that characterize pathological variances between TD and nTD subtypes of PD with contemporary mechanistic circuitry theories of PD symptom genesis.

Methods

We conducted and reported this study adhering to the PRISMA-ScR (PRISMA Extension for Scoping Reviews) guidelines and following the scoping review framework developed by Arksey and O’Malley and advanced by Levac et al. (Arksey and O'Malley 2005; Levac et al. 2010), a six-stage methodological framework involving (1) identifying the research question, (2) searching for relevant studies, (3) selecting studies, (4) charting the data, (5) collating, summarizing, and reporting the results, and (6) consulting with stakeholders.

Identifying relevant studies

A PubMed string search was iteratively developed with keywords “Parkinson's disease”, “tremor-dominant”, and “neuropathology” and variations thereof. The final string was used to identify all English-language abstracts containing human pathological data comparing the TD subtype to nTD subtype(s) within the NBIC database published up to December 2020. Additional articles were found from manual collation and supplemented references. The search string is provided in the Supplementary Material.

Selecting studies and charting the data

Eligibility was assessed via abstract and full-text screenings. Articles containing human pathological data comparing the TD PD subtype to the nTD subtype(s) directly were taken for further review. Data from all eligible studies pertaining to the tremor dominant, non-tremor dominant, and healthy control sample sizes, methodology used during pathological data collection, and conclusionary results were extracted and tabled.

Collating, summarizing, and reporting results

Results are grouped in the text by cardinal PD hallmarks including the dysfunction of the substantia nigra (SN) and dopaminergic system and the aggregation of Lewy bodies and amyloid-β plaques. We first discuss basal ganglia circuitry and the data, then describe how results coincide with neuroimaging research and fit into circuitopathy theories, as well as future clinical implications. While critical appraisal of studies is not required for scoping reviews, the main source of bias we identified were potential false positives stemming from small sample sizes. Additional plausible sources of bias include differential use of the MDS-UPDRS; practitioner- and patient-reported outcomes may vary between consolidated studies.

Consulting with relevant stakeholders

It is recommended under the scoping review guidelines to involve stakeholders throughout the review process (Levac et al. 2010). During the design and implementation, our project team regularly discussed the review strategy and our interpretations with experts in various clinical, technical, and academic fields including with authors of the reviewed studies. While there were no patients involved with this review, results will be shared with any patients involved in future clinical exercises supported from this work.

The substantia nigra

The SN is a bilateral nucleus in the basal ganglia (BG) and plays an important role in modulating movement. As the SN is a major source of dopamine, degeneration of dopamine neurons in the SN is the main pathological hallmark of PD (Alexander 2004). The SN can be divided into two subsections, the pars reticulata (SNr) that predominantly contains gamma-aminobutyric acid (GABA)-ergic neurons, mainly serving as an output nucleus in the cortico-basal ganglia loop conveying signals to the thalamus, superior colliculus, and to midbrain motor nuclei, and the pars compacta (SNc) projecting along the nigrostriatal pathway to the dorsal striatum (i.e., the caudate nucleus and putamen). SNc subdivisions innervate ventrally to the adjacent SNr (Haber and Knutson 2010) and modulate dopaminergic activity via axon collaterals. Fibers in the SNc have been shown to travel on average in a superior-posterior direction, whereas those in the SNr travel more in superior-medial/inferior-lateral directions (Plantinga et al. 2016). Medial, dorsal, and ventral SN dopaminergic regions generally project to the central and dorsal caudate, while caudal, lateral, and ventrolateral SN regions project more to the putamen (Bernheimer et al. 1973; Haber and Knutson 2010; Halliday et al. 1996; Jellinger 1999; Rinne et al. 1989).
The ventral region of the SNc is more vulnerable in numerous neurodegenerative diseases compared to the dorsal region partly due to the ventral tier being less melanised; vulnerability is concentrated ventrolaterally in PD and ventromedially in progressive supranuclear palsy (PSP) (Gibb and Lees 1991; Halliday et al. 1996). Functional neuroimaging has identified a tripartite connectivity-based parcellation of the SN; the medial SN functionally correlated to limbic striatal and cortical regions, the ventral SN to associative regions of cortex and striatum, and the lateral SN to somatomotor regions of striatum and cortex (Zhang et al. 2017). The medial SN could overlap with ventral tegmental area (VTA) as the VTA similarly projects via mesolimbic and mesocortical pathways to limbic and cortical areas. Due to a lack of clear tissue boundaries, this is sometimes referred to as the ‘SN/VTA’ complex.

Parkinsonian circuitry models; direct, indirect, hyperdirect pathways

Motor circuitry organization of BG pathways include the direct pathway (striatum-SNr/globus pallidus interna (GPi)-thalamus-cortex, responsible for motor excitation), the indirect pathway (striatum-globus pallidus externa (GPe)-subthalamic nucleus (STN)-SNr/GPi-thalamus-cortex, for motor inhibition), the hyperdirect pathway (STN-SNr/GPi-thalamus-cortex, as baseline motor inhibition), and the nigrostriatal projection (SNc-striatum, that is modulatory) (Neumann et al. 2018). The striatum is the main input for the SNr via the direct (striatum-SNr) and indirect (striatum-GPe-STN-SNr) pathways. However, the direct and indirect pathways are neurochemically distinct; direct pathway neurons express high levels of DA subtype-1 receptor (D1) while neurons in the indirect pathway express more D2 receptors (Cazorla et al. 2015). Because of this, striatal activity via the direct pathways has an inhibitory effect on the SNr, while indirect pathway activity has an excitatory effect; a decrease in striatal DA leads to hyperactivity in D2 containing neurons and hypoactivity in D1 neurons (Cazorla et al. 2015). Therefore, it was generally thought that hypotonic-hyperkinetic symptoms of PD such as tremors are caused by dysfunction of the indirect loop, leading to the loss of movement inhibition, while hypertonic-hypokinetic symptoms such as bradykinesia (a slowing of movement) are caused by dysfunction of the direct pathway that typically serves to plan and execute movements (Obeso et al. 2000), but neuropathological evidence for this is inconclusive.
Two classical functional hypotheses explaining the pathophysiological circuitry underlying movement dysfunction in PD include the ‘firing rate’ model and the ‘firing pattern’ model (Nambu et al. 2015). In the ‘firing rate’ model, dopamine depletion in the BG reduces excitation to striatal direct pathways that project to the GPi and reduces tonic inhibition to striatal indirect pathways that project to the GPe. This leads to an increase in the firing rates of the GPi and SNr via the inhibition of the striato-GPi/SNr direct pathway, excitation of the striato-GPe-STN-GPi/SNr indirect pathway, and subsequent decreased thalamic and cortical activity resulting in akinesia. A key limitation of the ‘firing rate’ model is that it does not explain tremor circuitries. The ‘firing pattern’ model suggests oscillatory and/or synchronized firing patterns in the BG disables the processing and relaying of motor-related information leading to movement dysfunction. These abnormal firing patterns exist in the form of bursts (a series of firings in short time periods) and oscillations (periodic bursts, ranging from 4 to 9 Hz ‘tremor frequency’ and 10 to 30 Hz ‘beta frequency’) (Ashkan et al. 2017). It is thought that dopamine depletion could enhance reciprocal connections between the GPe and the STN and lead to increased synchronized oscillatory activity resulting in motor dysfunction (Hutchison et al. 2004). While the ‘firing pattern’ model partially explains tremors, its explanation for akinesia is less clear.
Nambu et al. (2015) proposed a novel ‘dynamic activity’ model to reconcile the firing rate and firing pattern models, where signals from the direct pathway disinhibit thalamic neurons in a center thalamic area, hyperdirect and indirect pathways inhibit the center area, and hyperdirect and direct pathways inhibit the surround thalamic area (Fig. 1) (Nambu et al. 2015). The center-surround composition is based on an anatomical study showing excitatory STN–GPi fibers arborize more extensively and terminate more proximally in the thalamus than inhibitory striato–GPi fibers (Parent and Hazrati 1995), although the authors acknowledge that different studies suggest highly specific STN–GPi and striato-GPi projections (Smith et al. 1998; Shink et al. 1996). In this model, dopamine depletion is thought to reduce GPi inhibition via the direct pathway to the center area and facilitate GPi excitation via the hyperdirect and indirect pathways to the center and surround areas, leading to a reduction of thalamic and cortical disinhibition and subsequent akinesia. Furthermore, hyperkinetic disorders like tremors are said to result from the combination of enhanced inhibition to the center area of the GPi via the direct pathway and reduced excitation via the hyperdirect and indirect pathways to the center and surround area causing uncontrolled thalamic and cortical disinhibition. Further electrophysiological and anatomical studies investigating movement-related BG activity are needed to verify the ‘dynamic activity’ model. Moreover, how specific pathological differences between PD motor subtypes correspond to circuitry theories are prominently missing from all models.

Results

Basal ganglia degeneration between subtypes

An array of quantitative histological differences in basal nuclei degeneration have been shown between PD motor subtypes (Table 1). Histological research examining extensive clinical documentation of 45 autopsy confirmed PD patients with AR subtype show higher neuronal loss in the medial and lateral SN (devised by a line through the cerebral aqueduct) and the locus coeruleus (LC) compared to TD, as well as more severe SN gliosis, extraneuronal melanin deposits, and neuroaxonal dystrophy (Paulus and Jellinger 1991). In this study, neuroaxonal dystrophy of the SNr was seen in 10 out of 28 AR patients but in none of the 15 TD patients, while neuronal reduction in the dorsal raphe nucleus (DRN) was equal between subtypes (Paulus and Jellinger 1991).
Table 1
Neuropathological variances in Parkinson’s disease motor subtypes
Article
Patient numbers per subtype
Methods
Findings
PD
TD
nTD
PIGD
AR
Mixed
HC
Bernheimer et al. (1973)
28
     
28
IHC
Degree of akinesia correlated with DA and HVA decreases in the caudate nucleus, while the degree of tremor matched best with the degree of HVA decrease in the pallidum
Rinne et al. (1989)
12
     
18
IHC
Positive association found between rigidity and hypokinesia and low neuronal densities in the SNL
Paulus et al. (1991)
 
18
  
27
  
IHC
AR had a higher degree of gliosis in the SNM and SNL and more intense extraneuronal melanin deposits in the SNL. Neuroaxonal dystrophy of the SN reticula was seen in 10/28 AR but not in any of the 15 TD. AR have higher neuronal loss in the SNL and SNM and LC. Neuronal reduction in the DRN did not differ between AR and TD
Jellinger and Paulus (1992)
 
18
  
27
  
IHC
AR show degeneration of the striatonigral dopaminergic system and neuronal loss in SNL, SNM, and noradrencrgic LC. Tremor was related to other anatomo-pathophysiological substrates
Rajput et al. (1993)
 
34
 
11
   
x
The majority of tremor-onset cases had Lewy body disease while the majority of PIGD-onset cases had other forms of pathology
Halliday et al. (1996)
    
20
 
15
IHC
Greater dorsomedial SN cell loss was seen in PD patients with resting tremor than in those without tremor (85% loss compared to 70%, respectively)
McRitchie et al. (1997)
    
7
 
5
IHC
A8 regions appear largely unaffected in AR PD
Rajput et al. (2008)
 
3
  
3
2
5
HPLC
AR have greater loss of DA in both the dorsal and ventral regions of the rostral GPi compared to TD and Mixed
Selikhova et al. (2009)
242
88
93
    
Review
nTD have higher mean pathological grading of cortical Lewy bodies, more cortical amyloid-b plaque load, and more cerebral amyloid angiopathy compared to the EDO onset group and to TD. nTD have higher burdens of cortical Lewy bodies in the frontal regions and more severe plague formation in the neocortex compared to EOD and TD. EOD and TD were more likely to have brainstem and limbic Lewy body disease compared to nTD. Neocortical Lewy body class was more related to sever bradykinesia and falls than localized or limbic Lewy body disease was
Selikhova et al. (2013)
15
15 BT PD
     
IHC
Benign tremulous PD had less global neuronal loss in the substantia nigra compared to pathological controls. Benign tremulous PD had less cell loss in all nigral subregions compared to pathological controls. The most severe cell loss was seen in the ventrolateral nigra, while the medial nigra showed the greatest difference between benign tremulous PD and pathological controls
De Pablo-Fernandez et al. (2019)
111
      
Review
Staging of Lewy pathology and Alzheimer disease–related pathology did not differ between unique PD subtypes, but showed different rates of progression
A8 dopaminergic cell group A8, AR akinetic-rigid, BT benign tremulous, DA dopamine, DRN dorsal raphe nucleus, EOD early onset disease, GPi global pallidus internal, HC healthy control, HPLC high-performance liquid chromatography, HVA homovanillic acid, IHC immunohistochemistry, LC locus coeruleus, Mixed mixed-ratio subtype, nTD non-tremor dominant, PD Parkinson’s disease, PIGD postural instability and gait difficulty, SNL substantia nigra lateral, SN substantia nigra, SNM substantia nigra medial, TD tremor-dominant
In another study on pathologically confirmed PD patients with severe Parkinsonian AR syndrome, examinations showed that cell loss in the dorsolateral SN correlated with the duration of PD symptoms, while cell loss in the dorsomedial SN correlated with the presence of tremor (Halliday et al. 1996). TD showed more neuronal loss in the medial rather than in the lateral SNc, an area that projects to the caudate nucleus and anterior putamen (Jellinger 1999). The ventrolateral part of the SNc typically degenerates faster in AR PD relative to the medial SNc, with negative correlations between SNc cell counts, severity of AR symptoms, and dopamine loss in the posterior putamen (Jellinger 1999). When quantifying neuronal densities from the medial to lateral part of the SN in idiopathic PD brains and controls, a positive association was seen between rigidity and hypokinesia and low neuronal densities in the lateral SN, but tremor was less severe in patients with similarly low neuronal densities (Rinne et al. 1989). As the SN is the neuronal region where the enzyme tyrosine hydroxylase (TH) is most abundant, TH deficiency has shown to be associated with hypokinetic-rigid symptoms while other features like tremor are largely absent in patients with TH deficiency (Willemsen et al. 2010).
Although TD patients have shown less severe total SN cell loss (60%) compared to nTD (68%), TD patients have more cell loss in the periretrorubal field A8 in the lateral reticular formation (Hirsch 1991). A8 is located in the midbrain reticular formation dorso-caudal laterally to the SN and projects to the dorsolateral striatum and ventromedial thalamus (Deutch et al. 1988). Interestingly, in a study of seven autopsy confirmed AR patients with resting tremor, the A8 region appeared to be largely unaffected, with equal volume and neuronal count between the PD cases (those with only nigrostriatal pathway degeneration), Parkinson plus syndromes (those with additional degeneration elsewhere), or healthy controls (McRitchie et al. 1997). The degree of PD akinesia has been shown to correlate with decreased dopamine and its inactive metabolite homovanillic acid in the caudate nucleus, while tremor severity correlates more with reduced homovanillic acid in the pallidum (Bernheimer et al. 1973). Additionally, using high-performance liquid chromatography, AR patients have shown to have a greater loss of dopamine in both the dorsal and ventral regions of the rostral GPi compared to TD and mixed subtypes (Rajput et al. 2008).
It is clear that quantitative differences in BG degeneration are visible on histological levels between PD motor subtypes. Pathological results indicate a multitude of nigrostriatal degenerative processes innate to PD are less severe in TD compared to nTD; nTD patients show more severe degeneration in the lateral parts of the SN, while TD patients show more alterations in medial SN regions.

Cortical Lewy bodies and amyloid-β plaques between subtypes

Parkinsonian diseases involve abnormal accumulation of α-synuclein proteins within neuronal, glial, and nerve fiber cells. The progressive aggregation of Lewy bodies (LB), Lewy neurites, and amyloid-β plaques (Aβ) are additional hallmark histological markers of PD (Alexander 2004). In 1 study of 70 autopsy-verified PD patients, the majority of tremor-onset cases (55.9%) had LB disease while the majority of PIGD-onset cases (73%) had other or additional forms of pathology including multiple system atrophy and PSP (Rajput et al. 1993). This study aimed to evaluate the mode of onset and prognosis in autopsy confirmed PD cases and showed PIGD manifestations in PD were not due to age of onset as previously thought, but resulted from dissimilar neuropathology.
nTD patients have shown higher mean pathological grading of cortical LB, more cortical Aβ load, and more cerebral amyloid angiopathy compared to early-onset disease (EOD, < 45 years of age) and TD patients (Selikhova et al. 2009). In this study, the age of death was equal between cohorts indicating the higher cortical LB seen in nTD were not effects of age. Additionally, nTD showed higher cortical LB in the frontal regions and more severe plaque formation in the neocortex compared to EOD and TD who were more likely to have brainstem and limbic LB disease. Neocortical LB class was also related more to severe bradykinesia and falls than localized or limbic LB disease was (Selikhova et al. 2009). In De Pablo-Fernández et al., the staging of Lewy pathology and Alzheimer’s disease-related pathology did not differ between PD subtypes, but showed different rates of progression (De Pablo-Fernandez et al. 2019). Although this study did not group according to TD and nTD subtypes, instead using mild-motor predominant, intermediate, and diffuse malignant groupings, 79% of those in the mild motor-predominant subtype were TD and 33% of the diffuse malignant subtype were PIGD, and it demonstrated that the speed of pathological accumulation in certain neuronal areas varies between PD subtypes (Fereshtehnejad et al. 2017).
From these studies, it appears that patients with the nTD PD subtypes suffer more from cortical LB depositions and carry additional diverse forms of pathologies while TD patients have more localized pathology in the brainstem and limbic LB disease. Using cluster analyses, the authors suspect that a spectrum exists between a SN origin and a multi-pathway diffuse neurodegenerative process that is coupled with individual variation in progression and pathological patterns (Fereshtehnejad et al. 2017). As PD pathology gradually progresses from the brainstem to higher cortical layers as the disease advances, nTD patients with comparable disease duration to TD patients may be demonstrating greater and faster degeneration along this specific network that is not simply due to age or different stages of the disease.

Neuroimaging findings support pathology

Neuroimaging studies examining nTD and TD patients support findings of pathology variance in the SN. In Table 2, an overview is provided. Diffusion tensor imaging (DTI) measures water molecule diffusion and directionality and is an indirect marker for white matter composition. PD patients show reduced diffusion measurements in the ventrolateral SN in keeping with dopaminergic cell loss (Vaillancourt et al. 2009). Reductions of the DTI measurement fractional anisotropy (FA, reflecting a combination of axonal density, fiber mixture, and density) in the SN and putamen correlate with increased UPDRS motor scores in PD patients (Zhan et al. 2012). DTI has also shown increased mean diffusivity and radial diffusivity relating to structural disintegration in the SN being driven largely by the PIGD subtype (Nagae et al. 2016). Additionally, using neuromelanin-sensitive magnetic resonance imaging (NM-MRI) to detect dopamine metabolism, researchers found NM contrast-to-noise ratio (CNR) values in the lateral SN to decrease linearly with PD progression, while PIGD patients show larger decreases in CNR values in the lateral SN compared to TD (Wang et al. 2021). PIGD have also shown more severe NM signal attenuation in the medial SN compared to TD with the medial SN ipsilateral to the most clinically affected side showing the greatest power to discriminate PD motor subtypes (Xiang et al. 2017).
Table 2
Substantia nigra neuroimaging findings in Parkinson’s disease motor subtypes
Article
Patient numbers per subtype
Methods
Findings
PD
TD
nTD
PIGD
AR
Mixed
HC
[44]
14
     
14
DTI
Reduced FA in PD patients SN was greater in the caudal region compared with the middle and rostral regions
[45]
12
     
20
DTI
Alterations of FA in the SN and putamen correlate with increased UPDRS motor scores
[46]
9
12
    
20
DTI
Reduced FA and increased MD and RD in the SN was largely driven by PIGD. Increased diffusivity in the globus pallidus correlated with disease stage and motor severity in PIGD
[48]
 
9
 
14
  
20
NM-MRI
PIGD have more severe signal attenuation in the medial SN of compared to TD, with the medial SN ipsilateral to the MAH having the greatest power to discriminate PD motor subtypes
[49]
70
     
20
MR
SN MR phase shifts are positively correlated with UPDRS-III and bradykinesia-rigidity subscores but not with tremor subscores
[50]
 
10
  
10
 
20
MR
Positive correlation between motor subtype ratio and R2* in the putamen, caudate, and thalamus but not in SN, with larger TD ratios having higher R2*
DTI diffusion tensor imaging, HC healthy control, Mixed mixed-ratio subtype, nTD non-tremor dominant, PD Parkinson’s disease, PIGD postural instability and gait difficulty, SN substantia nigra, TD tremor-dominant, MD mean diffusivity, RD radial diffusivity, MAH most affected hemisphere, MR magnetic resonance imaging, NM-MRI neuromelanin-sensitive magnetic resonance imaging, R2* relaxometry rate, FA fractional anisotropy, UPDRS Unified Parkinson’s Disease Rating Scale
High-pass filtered phase MRI used to measure iron depositions show SN phase shifts to be positively correlated with UPDRS-III and bradykinesia-rigidity subscores but not with tremor subscores (Martin-Bastida et al. 2017). Additionally, MRI has found positive correlations between motor subtype ratio and measures of iron via transverse relaxation rates (R2*) in the putamen, caudate, and thalamus but not in the SN with larger TD motor phenotype ratios having higher R2* suggesting putaminal over nigral iron accumulation as an early TD predictor (Bunzeck et al. 2013). Taken together, these results suggest that regional patterns of microstructural degradation found via various neuroimaging techniques in the SN prove to accurately distinguish between PD motor subtypes and support histological findings assessing PD-related neurodegeneration. In short, neuroimaging shows nTD subtypes to have more laterally concentrated SN related severities compared to TD. Production of an accurate and detailed description of complete SN anatomy based on histological and immunohistochemistry stainings (e.g., Perl, Luxol fast blue/Cresyl violet, substance P, and Calbindin) of PD tissue conjointly imaged with high-field MRI (i.e., MR microscopy) has proven successful (Massey et al. 2017) warranting similar examination across PD subtypes.
How cortical neuritic plaque differences between PD subtypes correspond to neuroimaging data is less clear. Although MRI typically shows more severe cortical volumetric changes in PIGD compared to TD, on a whole volumetric variations between PD motor subtypes are mixed (Boonstra et al. 2021); TD patients have shown to have higher gray matter volumes (GMV) (Al-Bachari et al. 2017; Benninger et al. 2009; Herb et al. 2016; Piccinin et al. 2017), lower GMV (Rosenberg-Katz et al. 2013, 2016), and similar GMV (Karunanayaka et al. 2016; Linder et al. 2009; Nyberg et al. 2015; Prodoehl et al. 2013; Tessa et al. 2008; Vervoort et al. 2016) in cerebellular and cortical regions when compared to nTD subtypes. Correspondingly, greater attention should be granted towards cerebellular and brainstem pathways, as heterogeneous imaging acquisitions applied to all PD patients may lead to critical subcortical variations being overlooked. Parkinsonian resting tremor has shown to be related to cerebellar receiving thalamus activity (Helmich et al. 2012), although to our knowledge, no histological work has been done to support various neuroimaging studies that investigated cerebellar circuitopathies. This exact line of research will be a focus in our departments’ future work.

Pathology supports circuitry models

Evidence from pathological research does not definitively support the notion that tremor symptoms of PD are mainly caused by dysfunction of the indirect loop while bradykinesia is caused by dysfunction of the direct pathway. nTD patients show more severe cell loss in the ventrolateral part of the SN that projects to the dorsal putamen, while SNc cell loss has also shown to negatively correlate to severity of akinesia-rigidity and DA loss in the posterior putamen (Jellinger 1999). The ventrolateral SNc in nTD has additionally shown to degenerate more severely than the medial SN (Bernheimer et al. 1973) in contrast to TD patients, who exhibit less severe total and lateral SNc cell loss, but more severe cell loss in the medial SNc, as well as damage to the A8 region not seen in AR/nTD (Damier et al. 1999; Deutch et al. 1988; Jellinger and Paulus 1992).
nTD SN deficits could underpin pathological hyperactivity of the GABAergic indirect motor loop. GABAergic projections of the putamen have an inhibitory effect on the thalamus providing neuronal deficits in SN areas that project to the putamen to cause pathological thalamocortical pathway inhibition leading to reduced cortical activation. TD patients show pathological deficits in matrices of the dorsolateral striatum and ventromedial thalamus direct motor loop that could relate to hyperactivity in thalamomotor and cerebellar projections underpinning tremors. Such differential degeneration seen between PD motor subtypes could prove to be anatomical support for the Nambu et al., 2015‘dynamic activity’ model in how the GPi/SNr receives differential and competitive inputs from the hyperdirect, direct, and indirect pathways that modulate movement (Nambu et al. 2015). Variations within the proposed center-surround topographical distribution of striatum-derived inhibition on the GPi/SNr (Nambu et al. 2015) and its functional consequences could partly be explained by dissimilar neuropathological depositions in subpopulations of dopamine cells in the SN. nTD deficits in the ventrolateral SN could partly explain the proposed reduction in GPi/SNr inhibition in the center area and facilitation of GPi/SNr excitation in the center and surround areas that are suspected to lead to akinesia. Likewise, TD deficits in the medial SN could enhance inhibition in the center area of the GPi/SNr and reduced GPi/SNr excitation in the center and surrounding areas said to lead to involuntary movements.
Further research is needed to verify topographical distributions of SN subregion pathology. Investigations involving both subcortical and cortical stimulation are further required to functionally map striatum-derived inhibition and STN-derived excitation in the GPi to characterize ‘dynamic activity’ alterations between PD motor subtypes (Nambu et al. 2015). Tracing studies in non-human primates using cell-type specific viral tools (El-Shamayleh et al. 2016) can also shed light on projection patterns of medial and lateral SN dopamine cells. Additionally, how basal–brainstem pathways relate to diverse symptoms of PD remains to be studied in equal detail.

Clinical application

Modeling discrepancies between PD subtypes should have the ultimate aim to improve fundamental understandings of the disease and promote clinical success. Cortical pathological alternations could relate to the non-motor symptoms often seen more in nTD than in TD (Wu et al. 2016). In cognitively normal elderly individuals increased global amyloid pathology showed to be associated with alterations in gray matter networks indicative of network breakdowns that lead to dementia (Ten Kate et al. 2018). Although correlations of Aβ deposition with cognitive decline in PD is still a matter of debate (Compta et al. 2014; Shah et al. 2016; Melzer et al. 2019), a better understanding of variations in dopaminergic cell loss, plaque, and Lewy body depositions within PD patients has the potential to lead to more tailored treatment regimens and opens the opportunity to repurpose existing drug treatments, including dopamine (L-DOPA) based and amyloid targeted therapies (apomorphine) (Titova and Chaudhuri 2017).
Results hold further clinical implications in the field of deep brain stimulation (DBS) as TD patients tend to respond better to DBS, whereas those with axial subtypes and gait problems benefit less favorably (Neudorfer et al. 2019; Neumann et al. 2018; Xu et al. 2018) demonstrating a clear need for improved diagnoses between PD subtypes. The SN is being studied as a DBS target region for PD motor symptom alleviation (Sutton et al. 2013; Valldeoriola et al. 2019). Low-frequency SNr-DBS has shown to result in a significant improvement in PD patient’s freezing of gait, although its global antiparkinsonian effects were lower than that of the more common high frequency STN-DBS (Valldeoriola et al. 2019). STN-DBS has shown to have a protective effect on SNc neuron apoptosis in PD animal models conceivably resulting from neurotransmitter distribution and metabolism alterations (Wu et al. 2012), while supportive clinical data remain sparse.

Clinical limitation

It should be noted that the PD motor subtypes of TD and nTD have shown to have substantial clinical variability over time, specifically for the PIGD subtype (Simuni et al. 2016). After a 4-year follow-up in 325 PD patients, the proportion of recorded PIGD patients was shown to have increased significantly, from 23.1% at baseline to 44.2%. (Lee et al. 2019). Controversies exist that TD and PIGD are not different PD subtypes but instead are different clinical PD stages with dissimilar disease progression features (Lee et al. 2019). Correspondingly, the positive correlation between SN neurons and tremor may have been due to patients having not yet shifted from a TD subtype to nTD (Rinne et al. 1989). It is then reasonable to question if conventional PD motor subtyping has proficient clinical applicability. While authors found that 79% of those in the mild motor-predominant subtype were TD and 33% of the diffuse malignant subtype were PIGD, the tremor/PIGD classifications alone could not predict prognosis unlike their global subtyping solution that integrated additional daily living and cognition factors (Fereshtehnejad et al. 2017). These results speak towards the utility of incorporating various motor and non-motor symptoms as well as other biomarkers in the identification of PD subtypes, as more multivariate groupings may produce greater clinical applications and prove more stable over time (Ren et al. 2021). It is still unknown why certain subpopulations of cells undergo different degeneration patterns within each PD subtype, what initiates these divergences, whether they are a cause or a consequence of other co-occurring pathogenic mechanisms, and how they may be best targeted for treatment.

Conclusion

The purpose of scoping reviews is to chart a field within research and aid practitioners in constructing future research agendas (Arksey and O'Malley 2005). Clustering pathological differences between PD motor subtypes by combining MRI and histology to understand neuroanatomical pathways underlying clinical symptoms is essential in clarifying PD etiology. Literature displays a vast array of SN variation across PD subtypes, showing nTD to have more severe damage in the ventrolateral part of the SN while TD patients show less severe total SN cell loss but more severe medial SN alterations (Damier et al. 1999; Deutch et al. 1988; Jellinger and Paulus 1992). Patients with the nTD subtype further suffer from more cortical LB depositions and additional pathologies while TD patients have more brainstem and limbic LB disease (Rajput et al. 1993; Selikhova et al. 2009).
Neuroimaging studies support pathological findings between PD subtypes showing alterations within SN subsections (Martin-Bastida et al. 2017; Wang et al. 2021; Xiang et al. 2017), although support for cortical plaque depositions is less clear (Boonstra et al. 2021). Pathological differences between PD subtypes in the SN support the ‘dynamic activity’ model of PD symptom genesis; SN pathology may sustain the proposed center-surround topographical distribution of striatum-derived inhibition on the GPi/SNr that is correlated to motor disabilities (Nambu et al. 2015). Findings hold clinical implications in support of pathocircuitries and towards novel targets for treatment and interventions (Sutton et al. 2013; Valldeoriola et al. 2019; Wu et al. 2012).
Translational neuroscience works to disentangle the relationship between pathological data found ex vivo and clinically relevant in vivo data to innovate towards earlier PD diagnostic tools, better monitoring, and improved treatment approaches. Further research should continue to improve upon the creation of sustainable multivariate PD subtypes. As histology confers specificity by virtue of resolution neuroimaging should be inspired by postmortem findings and direct examinations with greater detail towards distinct neuronal pathway deficits.

Acknowledgements

The funding sources for study comes from a Stichting Beheer Vermogen De Weijerhorst. The authors would like to thank Geertjan van Zonneveld for his graphic design of Fig. 2.

Declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Ethical approval

This was a literature review where ethical approval from individuals was not required.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
Zurück zum Zitat Al-Bachari S, Vidyasagar R, Emsley HC, Parkes LM (2017) Structural and physiological neurovascular changes in idiopathic Parkinson’s disease and its clinical phenotypes. J Cereb Blood Flow Metab 37:3409–3421PubMedPubMedCentralCrossRef Al-Bachari S, Vidyasagar R, Emsley HC, Parkes LM (2017) Structural and physiological neurovascular changes in idiopathic Parkinson’s disease and its clinical phenotypes. J Cereb Blood Flow Metab 37:3409–3421PubMedPubMedCentralCrossRef
Zurück zum Zitat Alexander GE (2004) Biology of Parkinson’s disease: pathogenesis and pathophysiology of a multisystem neurodegenerative disorder. Dialogues Clin Neurosci 6:259–280PubMedPubMedCentralCrossRef Alexander GE (2004) Biology of Parkinson’s disease: pathogenesis and pathophysiology of a multisystem neurodegenerative disorder. Dialogues Clin Neurosci 6:259–280PubMedPubMedCentralCrossRef
Zurück zum Zitat Alkemade A, Mulder MJ, Groot JM, Isaacs BR, van Berendonk N, Lute N, Isherwood SJ, Bazin PL, Forstmann BU (2020) The Amsterdam Ultra-high field adult lifespan database (AHEAD): a freely available multimodal 7 Tesla submillimeter magnetic resonance imaging database. Neuroimage 221:117200PubMedCrossRef Alkemade A, Mulder MJ, Groot JM, Isaacs BR, van Berendonk N, Lute N, Isherwood SJ, Bazin PL, Forstmann BU (2020) The Amsterdam Ultra-high field adult lifespan database (AHEAD): a freely available multimodal 7 Tesla submillimeter magnetic resonance imaging database. Neuroimage 221:117200PubMedCrossRef
Zurück zum Zitat Arksey H, O’Malley L (2005) Scoping studies: towards a methodological framework. Int J Soc Res Methodol 8:19–32CrossRef Arksey H, O’Malley L (2005) Scoping studies: towards a methodological framework. Int J Soc Res Methodol 8:19–32CrossRef
Zurück zum Zitat Ashkan K, Rogers P, Bergman H, Ughratdar I (2017) Insights into the mechanisms of deep brain stimulation. Nat Rev Neurol 13:548–554PubMedCrossRef Ashkan K, Rogers P, Bergman H, Ughratdar I (2017) Insights into the mechanisms of deep brain stimulation. Nat Rev Neurol 13:548–554PubMedCrossRef
Zurück zum Zitat Benninger DH, Thees S, Kollias SS, Bassetti CL, Waldvogel D (2009) Morphological differences in Parkinson’s disease with and without rest tremor. J Neurol 256:256–263PubMedCrossRef Benninger DH, Thees S, Kollias SS, Bassetti CL, Waldvogel D (2009) Morphological differences in Parkinson’s disease with and without rest tremor. J Neurol 256:256–263PubMedCrossRef
Zurück zum Zitat Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F (1973) Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci 20:415–455PubMedCrossRef Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F (1973) Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci 20:415–455PubMedCrossRef
Zurück zum Zitat Boonstra JT, Michielse S, Temel Y, Hoogland G, Jahanshahi A (2021) Neuroimaging detectable differences between Parkinson’s disease motor subtypes: a systematic review. Mov Disord Clin Pract 8:175–192PubMedCrossRef Boonstra JT, Michielse S, Temel Y, Hoogland G, Jahanshahi A (2021) Neuroimaging detectable differences between Parkinson’s disease motor subtypes: a systematic review. Mov Disord Clin Pract 8:175–192PubMedCrossRef
Zurück zum Zitat Borghammer P (2018) How does Parkinson’s disease begin? Perspectives on neuroanatomical pathways, prions, and histology. Mov Disord 33:48–57PubMedCrossRef Borghammer P (2018) How does Parkinson’s disease begin? Perspectives on neuroanatomical pathways, prions, and histology. Mov Disord 33:48–57PubMedCrossRef
Zurück zum Zitat Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E (2003) Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 24:197–211PubMedCrossRef Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E (2003) Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging 24:197–211PubMedCrossRef
Zurück zum Zitat Bunzeck N, Singh-Curry V, Eckart C, Weiskopf N, Perry RJ, Bain PG, Duzel E, Husain M (2013) Motor phenotype and magnetic resonance measures of basal ganglia iron levels in Parkinson’s disease. Parkinsonism Relat Disord 19:1136–1142PubMedPubMedCentralCrossRef Bunzeck N, Singh-Curry V, Eckart C, Weiskopf N, Perry RJ, Bain PG, Duzel E, Husain M (2013) Motor phenotype and magnetic resonance measures of basal ganglia iron levels in Parkinson’s disease. Parkinsonism Relat Disord 19:1136–1142PubMedPubMedCentralCrossRef
Zurück zum Zitat Cazorla M, Kang UJ, Kellendonk C (2015) Balancing the basal ganglia circuitry: a possible new role for dopamine D2 receptors in health and disease. Mov Disord 30:895–903PubMedPubMedCentralCrossRef Cazorla M, Kang UJ, Kellendonk C (2015) Balancing the basal ganglia circuitry: a possible new role for dopamine D2 receptors in health and disease. Mov Disord 30:895–903PubMedPubMedCentralCrossRef
Zurück zum Zitat Compta Y, Parkkinen L, Kempster P, Selikhova M, Lashley T, Holton JL, Lees AJ, Revesz T (2014) The significance of alpha-synuclein, amyloid-beta and tau pathologies in Parkinson’s disease progression and related dementia. Neurodegener Dis 13:154–156PubMedCrossRef Compta Y, Parkkinen L, Kempster P, Selikhova M, Lashley T, Holton JL, Lees AJ, Revesz T (2014) The significance of alpha-synuclein, amyloid-beta and tau pathologies in Parkinson’s disease progression and related dementia. Neurodegener Dis 13:154–156PubMedCrossRef
Zurück zum Zitat Damier P, Hirsch EC, Agid Y, Graybiel AM (1999) The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain 122(Pt 8):1437–1448PubMedCrossRef Damier P, Hirsch EC, Agid Y, Graybiel AM (1999) The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain 122(Pt 8):1437–1448PubMedCrossRef
Zurück zum Zitat De Pablo-Fernandez E, Lees AJ, Holton JL, Warner TT (2019) Prognosis and neuropathologic correlation of clinical subtypes of Parkinson disease. JAMA Neurol 76:470–479PubMedPubMedCentralCrossRef De Pablo-Fernandez E, Lees AJ, Holton JL, Warner TT (2019) Prognosis and neuropathologic correlation of clinical subtypes of Parkinson disease. JAMA Neurol 76:470–479PubMedPubMedCentralCrossRef
Zurück zum Zitat Deutch AY, Goldstein M, Baldino F Jr, Roth RH (1988) Telencephalic projections of the A8 dopamine cell group. Ann N Y Acad Sci 537:27–50PubMedCrossRef Deutch AY, Goldstein M, Baldino F Jr, Roth RH (1988) Telencephalic projections of the A8 dopamine cell group. Ann N Y Acad Sci 537:27–50PubMedCrossRef
Zurück zum Zitat Fereshtehnejad SM, Zeighami Y, Dagher A, Postuma RB (2017) Clinical criteria for subtyping Parkinson’s disease: biomarkers and longitudinal progression. Brain 140:1959–1976PubMedCrossRef Fereshtehnejad SM, Zeighami Y, Dagher A, Postuma RB (2017) Clinical criteria for subtyping Parkinson’s disease: biomarkers and longitudinal progression. Brain 140:1959–1976PubMedCrossRef
Zurück zum Zitat Gibb WR, Lees AJ (1991) Anatomy, pigmentation, ventral and dorsal subpopulations of the substantia nigra, and differential cell death in Parkinson’s disease. J Neurol Neurosurg Psychiatry 54:388–396PubMedPubMedCentralCrossRef Gibb WR, Lees AJ (1991) Anatomy, pigmentation, ventral and dorsal subpopulations of the substantia nigra, and differential cell death in Parkinson’s disease. J Neurol Neurosurg Psychiatry 54:388–396PubMedPubMedCentralCrossRef
Zurück zum Zitat Goetz CG, Fahn S, Martinez-Martin P, Poewe W, Sampaio C, Stebbins GT, Stern MB, Tilley BC, Dodel R, Dubois B, Holloway R, Jankovic J, Kulisevsky J, Lang AE, Lees A, Leurgans S, LeWitt PA, Nyenhuis D, Olanow CW, Rascol O, Schrag A, Teresi JA, Van Hilten JJ, LaPelle N (2007) Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): process, format, and clinimetric testing plan. Mov Disord 22:41–47PubMedCrossRef Goetz CG, Fahn S, Martinez-Martin P, Poewe W, Sampaio C, Stebbins GT, Stern MB, Tilley BC, Dodel R, Dubois B, Holloway R, Jankovic J, Kulisevsky J, Lang AE, Lees A, Leurgans S, LeWitt PA, Nyenhuis D, Olanow CW, Rascol O, Schrag A, Teresi JA, Van Hilten JJ, LaPelle N (2007) Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): process, format, and clinimetric testing plan. Mov Disord 22:41–47PubMedCrossRef
Zurück zum Zitat Haber SN, Knutson B (2010) The reward circuit: linking primate anatomy and human imaging. Neuropsychopharmacology 35:4–26PubMedCrossRef Haber SN, Knutson B (2010) The reward circuit: linking primate anatomy and human imaging. Neuropsychopharmacology 35:4–26PubMedCrossRef
Zurück zum Zitat Halliday GM, McRitchie DA, Cartwright H, Pamphlett R, Hely MA, Morris JG (1996) Midbrain neuropathology in idiopathic Parkinson’s disease and diffuse Lewy body disease. J Clin Neurosci 3:52–60PubMedCrossRef Halliday GM, McRitchie DA, Cartwright H, Pamphlett R, Hely MA, Morris JG (1996) Midbrain neuropathology in idiopathic Parkinson’s disease and diffuse Lewy body disease. J Clin Neurosci 3:52–60PubMedCrossRef
Zurück zum Zitat He N, Huang P, Ling H, Langley J, Liu C, Ding B, Huang J, Xu H, Zhang Y, Zhang Z, Hu X, Chen S, Yan F (2017) Dentate nucleus iron deposition is a potential biomarker for tremor-dominant Parkinson’s disease. NMR Biomed 30:e3554CrossRef He N, Huang P, Ling H, Langley J, Liu C, Ding B, Huang J, Xu H, Zhang Y, Zhang Z, Hu X, Chen S, Yan F (2017) Dentate nucleus iron deposition is a potential biomarker for tremor-dominant Parkinson’s disease. NMR Biomed 30:e3554CrossRef
Zurück zum Zitat Helmich RC, Hallett M, Deuschl G, Toni I, Bloem BR (2012) Cerebral causes and consequences of parkinsonian resting tremor: a tale of two circuits? Brain 135:3206–3226PubMedPubMedCentralCrossRef Helmich RC, Hallett M, Deuschl G, Toni I, Bloem BR (2012) Cerebral causes and consequences of parkinsonian resting tremor: a tale of two circuits? Brain 135:3206–3226PubMedPubMedCentralCrossRef
Zurück zum Zitat Herb JN, Rane S, Isaacs DA, Van Wouwe N, Roman OC, Landman BA, Dawant BM, Hedera P, Zald DH, Neimat JS, Wylie SA, Donahue MJ, Claassen DO (2016) Cortical implications of advancing age and disease duration in Parkinson’s disease patients with postural instability and gait dysfunction. J Parkinsons Dis 6:441–451PubMedPubMedCentralCrossRef Herb JN, Rane S, Isaacs DA, Van Wouwe N, Roman OC, Landman BA, Dawant BM, Hedera P, Zald DH, Neimat JS, Wylie SA, Donahue MJ, Claassen DO (2016) Cortical implications of advancing age and disease duration in Parkinson’s disease patients with postural instability and gait dysfunction. J Parkinsons Dis 6:441–451PubMedPubMedCentralCrossRef
Zurück zum Zitat Hirsch EC, Mouatt A, Graybiel AM, Javoy-Agid F, Agid Y (1991) The loss of catecholami-nergic neurons in Parkinson’s disease is heterogeneous in the midbrain. 10th international symposium on Parkinson’s disease. In: 10th International Symposium on Parkinson’s disease Hirsch EC, Mouatt A, Graybiel AM, Javoy-Agid F, Agid Y (1991) The loss of catecholami-nergic neurons in Parkinson’s disease is heterogeneous in the midbrain. 10th international symposium on Parkinson’s disease. In: 10th International Symposium on Parkinson’s disease
Zurück zum Zitat Hutchison WD, Dostrovsky JO, Walters JR, Courtemanche R, Boraud T, Goldberg J, Brown P (2004) Neuronal oscillations in the basal ganglia and movement disorders: evidence from whole animal and human recordings. J Neurosci 24:9240–9243PubMedPubMedCentralCrossRef Hutchison WD, Dostrovsky JO, Walters JR, Courtemanche R, Boraud T, Goldberg J, Brown P (2004) Neuronal oscillations in the basal ganglia and movement disorders: evidence from whole animal and human recordings. J Neurosci 24:9240–9243PubMedPubMedCentralCrossRef
Zurück zum Zitat Jankovic J, Carter J, Gauthier S, Goetz C, Golbe L, Huber S, Koller W, Olanow C, Shoulson I, Stern M, Tanner C, Weiner W (1990) Variable expression of Parkinson’s disease: a base-line analysis of the DATATOP cohort. Neurology 40:1529PubMedCrossRef Jankovic J, Carter J, Gauthier S, Goetz C, Golbe L, Huber S, Koller W, Olanow C, Shoulson I, Stern M, Tanner C, Weiner W (1990) Variable expression of Parkinson’s disease: a base-line analysis of the DATATOP cohort. Neurology 40:1529PubMedCrossRef
Zurück zum Zitat Jellinger K (1999) Post mortem studies in Parkinson’s disease—is it possible to detect brain areas for specific symptoms. Journal of neural transmission, supplement. Springer, Vienna Jellinger K (1999) Post mortem studies in Parkinson’s disease—is it possible to detect brain areas for specific symptoms. Journal of neural transmission, supplement. Springer, Vienna
Zurück zum Zitat Jellinger KA, Paulus W (1992) Clinico-pathological correlations in Parkinson’s disease. Clin Neurol Neurosurg 94(Suppl):S86–S88PubMedCrossRef Jellinger KA, Paulus W (1992) Clinico-pathological correlations in Parkinson’s disease. Clin Neurol Neurosurg 94(Suppl):S86–S88PubMedCrossRef
Zurück zum Zitat Karunanayaka PR, Lee EY, Lewis MM, Sen S, Eslinger PJ, Yang QX, Huang X (2016) Default mode network differences between rigidity- and tremor-predominant Parkinson’s disease. Cortex 81:239–250PubMedPubMedCentralCrossRef Karunanayaka PR, Lee EY, Lewis MM, Sen S, Eslinger PJ, Yang QX, Huang X (2016) Default mode network differences between rigidity- and tremor-predominant Parkinson’s disease. Cortex 81:239–250PubMedPubMedCentralCrossRef
Zurück zum Zitat Lee JW, Song YS, Kim H, Ku BD, Lee WW (2019) Alteration of tremor dominant and postural instability gait difficulty subtypes during the progression of Parkinson’s disease: analysis of the PPMI cohort. Front Neurol 10:471PubMedPubMedCentralCrossRef Lee JW, Song YS, Kim H, Ku BD, Lee WW (2019) Alteration of tremor dominant and postural instability gait difficulty subtypes during the progression of Parkinson’s disease: analysis of the PPMI cohort. Front Neurol 10:471PubMedPubMedCentralCrossRef
Zurück zum Zitat Linder J, Birgander R, Olsson I, Riklund K, Larsson AK, Edstrom M, Stenlund H, Forsgren L (2009) Degenerative changes were common in brain magnetic resonance imaging in patients with newly diagnosed Parkinson’s disease in a population-based cohort. J Neurol 256:1671–1680PubMedCrossRef Linder J, Birgander R, Olsson I, Riklund K, Larsson AK, Edstrom M, Stenlund H, Forsgren L (2009) Degenerative changes were common in brain magnetic resonance imaging in patients with newly diagnosed Parkinson’s disease in a population-based cohort. J Neurol 256:1671–1680PubMedCrossRef
Zurück zum Zitat Marras C, Beck JC, Bower JH, Roberts E, Ritz B, Ross GW, Abbott RD, Savica R, Van Den Eeden SK, Willis AW, Tanner CM, P. Group Parkinson’s Foundation (2018) Prevalence of Parkinson’s disease across North America. NPJ Parkinsons Dis 4:21PubMedPubMedCentralCrossRef Marras C, Beck JC, Bower JH, Roberts E, Ritz B, Ross GW, Abbott RD, Savica R, Van Den Eeden SK, Willis AW, Tanner CM, P. Group Parkinson’s Foundation (2018) Prevalence of Parkinson’s disease across North America. NPJ Parkinsons Dis 4:21PubMedPubMedCentralCrossRef
Zurück zum Zitat 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 Parkinson’s disease: a cross-sectional study of iron-related magnetic resonance imaging susceptibility. Eur J Neurol 24:357–365PubMedCrossRef 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 Parkinson’s disease: a cross-sectional study of iron-related magnetic resonance imaging susceptibility. Eur J Neurol 24:357–365PubMedCrossRef
Zurück zum Zitat Massey LA, Miranda MA, Al-Helli O, Parkes HG, Thornton JS, So PW, White MJ, Mancini L, Strand C, Holton J, Lees AJ, Revesz T, Yousry TA (2017) 9.4 T MR microscopy of the substantia nigra with pathological validation in controls and disease. Neuroimage Clin 13:154–163PubMedCrossRef Massey LA, Miranda MA, Al-Helli O, Parkes HG, Thornton JS, So PW, White MJ, Mancini L, Strand C, Holton J, Lees AJ, Revesz T, Yousry TA (2017) 9.4 T MR microscopy of the substantia nigra with pathological validation in controls and disease. Neuroimage Clin 13:154–163PubMedCrossRef
Zurück zum Zitat McRitchie DA, Cartwright HR, Halliday GM (1997) Specific A10 dopaminergic nuclei in the midbrain degenerate in Parkinson’s disease. Exp Neurol 144:202–213PubMedCrossRef McRitchie DA, Cartwright HR, Halliday GM (1997) Specific A10 dopaminergic nuclei in the midbrain degenerate in Parkinson’s disease. Exp Neurol 144:202–213PubMedCrossRef
Zurück zum Zitat Melzer TR, Stark MR, Keenan RJ, Myall DJ, MacAskill MR, Pitcher TL, Livingston L, Grenfell S, Horne KL, Young BN, Pascoe MJ, Almuqbel MM, Wang J, Marsh SH, Miller DH, Dalrymple-Alford JC, Anderson TJ (2019) Beta amyloid deposition is not associated with cognitive impairment in Parkinson’s disease. Front Neurol 10:391PubMedPubMedCentralCrossRef Melzer TR, Stark MR, Keenan RJ, Myall DJ, MacAskill MR, Pitcher TL, Livingston L, Grenfell S, Horne KL, Young BN, Pascoe MJ, Almuqbel MM, Wang J, Marsh SH, Miller DH, Dalrymple-Alford JC, Anderson TJ (2019) Beta amyloid deposition is not associated with cognitive impairment in Parkinson’s disease. Front Neurol 10:391PubMedPubMedCentralCrossRef
Zurück zum Zitat Nagae LM, Honce JM, Tanabe J, Shelton E, Sillau SH, Berman BD (2016) Microstructural changes within the basal ganglia differ between Parkinson disease subtypes. Front Neuroanat 10:17PubMedPubMedCentralCrossRef Nagae LM, Honce JM, Tanabe J, Shelton E, Sillau SH, Berman BD (2016) Microstructural changes within the basal ganglia differ between Parkinson disease subtypes. Front Neuroanat 10:17PubMedPubMedCentralCrossRef
Zurück zum Zitat Nambu A, Tachibana Y, Chiken S (2015) Cause of parkinsonian symptoms: firing rate, firing pattern or dynamic activity changes? Basal Ganglia 5:1–6CrossRef Nambu A, Tachibana Y, Chiken S (2015) Cause of parkinsonian symptoms: firing rate, firing pattern or dynamic activity changes? Basal Ganglia 5:1–6CrossRef
Zurück zum Zitat Neudorfer C, Hinzke M, Hunsche S, El Majdoub F, Lozano A, Maarouf M (2019) Combined deep brain stimulation of subthalamic nucleus and ventral intermediate thalamic nucleus in tremor-dominant Parkinson’s disease using a parietal approach. Neuromodulation 22:493–502PubMedCrossRef Neudorfer C, Hinzke M, Hunsche S, El Majdoub F, Lozano A, Maarouf M (2019) Combined deep brain stimulation of subthalamic nucleus and ventral intermediate thalamic nucleus in tremor-dominant Parkinson’s disease using a parietal approach. Neuromodulation 22:493–502PubMedCrossRef
Zurück zum Zitat Neumann WJ, Schroll H, de Almeida Marcelino AL, Horn A, Ewert S, Irmen F, Krause P, Schneider GH, Hamker F, Kuhn AA (2018) Functional segregation of basal ganglia pathways in Parkinson’s disease. Brain 141:2655–2669PubMed Neumann WJ, Schroll H, de Almeida Marcelino AL, Horn A, Ewert S, Irmen F, Krause P, Schneider GH, Hamker F, Kuhn AA (2018) Functional segregation of basal ganglia pathways in Parkinson’s disease. Brain 141:2655–2669PubMed
Zurück zum Zitat Nyberg EM, Tanabe J, Honce JM, Krmpotich T, Shelton E, Hedeman J, Berman BD (2015) Morphologic changes in the mesolimbic pathway in Parkinson’s disease motor subtypes. Parkinsonism Relat Disord 21:536–540PubMedPubMedCentralCrossRef Nyberg EM, Tanabe J, Honce JM, Krmpotich T, Shelton E, Hedeman J, Berman BD (2015) Morphologic changes in the mesolimbic pathway in Parkinson’s disease motor subtypes. Parkinsonism Relat Disord 21:536–540PubMedPubMedCentralCrossRef
Zurück zum Zitat Obeso JA, Rodriguez-Oroz MC, Rodriguez M, Lanciego JL, Artieda J, Gonzalo N, Olanow CW (2000) Pathophysiology of the basal ganglia in Parkinson’s disease. Trends Neurosci 23:S8-19PubMedCrossRef Obeso JA, Rodriguez-Oroz MC, Rodriguez M, Lanciego JL, Artieda J, Gonzalo N, Olanow CW (2000) Pathophysiology of the basal ganglia in Parkinson’s disease. Trends Neurosci 23:S8-19PubMedCrossRef
Zurück zum Zitat Parent A, Hazrati LN (1995) Functional anatomy of the basal ganglia. II. The place of subthalamic nucleus and external pallidum in basal ganglia circuitry. Brain Res Brain Res Rev 20:128–154PubMedCrossRef Parent A, Hazrati LN (1995) Functional anatomy of the basal ganglia. II. The place of subthalamic nucleus and external pallidum in basal ganglia circuitry. Brain Res Brain Res Rev 20:128–154PubMedCrossRef
Zurück zum Zitat Paulus W, Jellinger K (1991) The neuropathologic basis of different clinical subgroups of Parkinson’s disease. J Neuropathol Exp Neurol 50:743–755PubMedCrossRef Paulus W, Jellinger K (1991) The neuropathologic basis of different clinical subgroups of Parkinson’s disease. J Neuropathol Exp Neurol 50:743–755PubMedCrossRef
Zurück zum Zitat Piccinin CC, Campos LS, Guimaraes RP, Piovesana LG, Dos Santos MCA, Azevedo PC, Campos BM, de Rezende TJR, Amato-Filho A, Cendes F, D’Abreu A (2017) Differential pattern of cerebellar atrophy in tremor-predominant and akinetic/rigidity-predominant Parkinson’s disease. Cerebellum 16:623–628PubMedCrossRef Piccinin CC, Campos LS, Guimaraes RP, Piovesana LG, Dos Santos MCA, Azevedo PC, Campos BM, de Rezende TJR, Amato-Filho A, Cendes F, D’Abreu A (2017) Differential pattern of cerebellar atrophy in tremor-predominant and akinetic/rigidity-predominant Parkinson’s disease. Cerebellum 16:623–628PubMedCrossRef
Zurück zum Zitat Plantinga BR, Roebroeck A, Kemper VG, Uludag K, Melse M, Mai J, Kuijf ML, Herrler A, Jahanshahi A, Ter Haar Romeny BM, Temel Y (2016) Ultra-high field MRI post mortem structural connectivity of the human subthalamic nucleus, substantia nigra, and globus pallidus. Front Neuroanat 10:66PubMedPubMedCentralCrossRef Plantinga BR, Roebroeck A, Kemper VG, Uludag K, Melse M, Mai J, Kuijf ML, Herrler A, Jahanshahi A, Ter Haar Romeny BM, Temel Y (2016) Ultra-high field MRI post mortem structural connectivity of the human subthalamic nucleus, substantia nigra, and globus pallidus. Front Neuroanat 10:66PubMedPubMedCentralCrossRef
Zurück zum Zitat Prodoehl J, Planetta PJ, Kurani AS, Comella CL, Corcos DM, Vaillancourt DE (2013) Differences in brain activation between tremor- and nontremor-dominant Parkinson disease. JAMA Neurol 70:100–106PubMedPubMedCentralCrossRef Prodoehl J, Planetta PJ, Kurani AS, Comella CL, Corcos DM, Vaillancourt DE (2013) Differences in brain activation between tremor- and nontremor-dominant Parkinson disease. JAMA Neurol 70:100–106PubMedPubMedCentralCrossRef
Zurück zum Zitat Rajput AH, Pahwa R, Pahwa P, Rajput A (1993) Prognostic significance of the onset mode in parkinsonism. Neurology 43:829–830PubMedCrossRef Rajput AH, Pahwa R, Pahwa P, Rajput A (1993) Prognostic significance of the onset mode in parkinsonism. Neurology 43:829–830PubMedCrossRef
Zurück zum Zitat Rajput AH, Sitte HH, Rajput A, Fenton ME, Pifl C, Hornykiewicz O (2008) Globus pallidus dopamine and Parkinson motor subtypes: clinical and brain biochemical correlation. Neurology 70:1403–1410PubMedCrossRef Rajput AH, Sitte HH, Rajput A, Fenton ME, Pifl C, Hornykiewicz O (2008) Globus pallidus dopamine and Parkinson motor subtypes: clinical and brain biochemical correlation. Neurology 70:1403–1410PubMedCrossRef
Zurück zum Zitat Rajput AH, Voll A, Rajput ML, Robinson CA, Rajput A (2009) Course in Parkinson disease subtypes: a 39-year clinicopathologic study. Neurology 73:206–212PubMedCrossRef Rajput AH, Voll A, Rajput ML, Robinson CA, Rajput A (2009) Course in Parkinson disease subtypes: a 39-year clinicopathologic study. Neurology 73:206–212PubMedCrossRef
Zurück zum Zitat Ren J, Pan C, Li Y, Li L, Hua P, Xu L, Zhang L, Zhang W, Xu P, Liu W (2021) Consistency and stability of motor subtype classifications in patients with de novo Parkinson’s disease. Front Neurosci 15:637896PubMedPubMedCentralCrossRef Ren J, Pan C, Li Y, Li L, Hua P, Xu L, Zhang L, Zhang W, Xu P, Liu W (2021) Consistency and stability of motor subtype classifications in patients with de novo Parkinson’s disease. Front Neurosci 15:637896PubMedPubMedCentralCrossRef
Zurück zum Zitat Rinne JO, Rummukainen J, Paljarvi L, Rinne UK (1989) Dementia in Parkinson’s disease is related to neuronal loss in the medial substantia nigra. Ann Neurol 26:47–50PubMedCrossRef Rinne JO, Rummukainen J, Paljarvi L, Rinne UK (1989) Dementia in Parkinson’s disease is related to neuronal loss in the medial substantia nigra. Ann Neurol 26:47–50PubMedCrossRef
Zurück zum Zitat Rosenberg-Katz K, Herman T, Jacob Y, Giladi N, Hendler T, Hausdorff JM (2013) Gray matter atrophy distinguishes between Parkinson disease motor subtypes. Neurology 80:1476–1484PubMedPubMedCentralCrossRef Rosenberg-Katz K, Herman T, Jacob Y, Giladi N, Hendler T, Hausdorff JM (2013) Gray matter atrophy distinguishes between Parkinson disease motor subtypes. Neurology 80:1476–1484PubMedPubMedCentralCrossRef
Zurück zum Zitat Rosenberg-Katz K, Herman T, Jacob Y, Kliper E, Giladi N, Hausdorff JM (2016) Subcortical volumes differ in Parkinson’s disease motor subtypes: new insights into the pathophysiology of disparate symptoms. Front Hum Neurosci 10:356PubMedPubMedCentralCrossRef Rosenberg-Katz K, Herman T, Jacob Y, Kliper E, Giladi N, Hausdorff JM (2016) Subcortical volumes differ in Parkinson’s disease motor subtypes: new insights into the pathophysiology of disparate symptoms. Front Hum Neurosci 10:356PubMedPubMedCentralCrossRef
Zurück zum Zitat Selikhova M, Williams DR, Kempster PA, Holton JL, Revesz T, Lees AJ (2009) A clinico-pathological study of subtypes in Parkinson’s disease. Brain 132:2947–2957PubMedCrossRef Selikhova M, Williams DR, Kempster PA, Holton JL, Revesz T, Lees AJ (2009) A clinico-pathological study of subtypes in Parkinson’s disease. Brain 132:2947–2957PubMedCrossRef
Zurück zum Zitat Shah N, Frey KA, Muller ML, Petrou M, Kotagal V, Koeppe RA, Scott PJ, Albin RL, Bohnen NI (2016) Striatal and cortical beta-amyloidopathy and cognition in Parkinson’s disease. Mov Disord 31:111–117PubMedCrossRef Shah N, Frey KA, Muller ML, Petrou M, Kotagal V, Koeppe RA, Scott PJ, Albin RL, Bohnen NI (2016) Striatal and cortical beta-amyloidopathy and cognition in Parkinson’s disease. Mov Disord 31:111–117PubMedCrossRef
Zurück zum Zitat Shink E, Bevan MD, Bolam JP, Smith Y (1996) The subthalamic nucleus and the external pallidum: two tightly interconnected structures that control the output of the basal ganglia in the monkey. Neuroscience 73:335–357PubMedCrossRef Shink E, Bevan MD, Bolam JP, Smith Y (1996) The subthalamic nucleus and the external pallidum: two tightly interconnected structures that control the output of the basal ganglia in the monkey. Neuroscience 73:335–357PubMedCrossRef
Zurück zum Zitat Simuni T, Caspell-Garcia C, Coffey C, Lasch S, Tanner C, Marek K, Investigators P (2016) How stable are Parkinson’s disease subtypes in de novo patients: analysis of the PPMI cohort? Parkinsonism Relat Disord 28:62–67PubMedCrossRef Simuni T, Caspell-Garcia C, Coffey C, Lasch S, Tanner C, Marek K, Investigators P (2016) How stable are Parkinson’s disease subtypes in de novo patients: analysis of the PPMI cohort? Parkinsonism Relat Disord 28:62–67PubMedCrossRef
Zurück zum Zitat Smith Y, Bevan MD, Shink E, Bolam JP (1998) Microcircuitry of the direct and indirect pathways of the basal ganglia. Neuroscience 86:353–387PubMed Smith Y, Bevan MD, Shink E, Bolam JP (1998) Microcircuitry of the direct and indirect pathways of the basal ganglia. Neuroscience 86:353–387PubMed
Zurück zum Zitat Sutton AC, Yu W, Calos ME, Smith AB, Ramirez-Zamora A, Molho ES, Pilitsis JG, Brotchie JM, Shin DS (2013) Deep brain stimulation of the substantia nigra pars reticulata improves forelimb akinesia in the hemiparkinsonian rat. J Neurophysiol 109:363–374PubMedCrossRef Sutton AC, Yu W, Calos ME, Smith AB, Ramirez-Zamora A, Molho ES, Pilitsis JG, Brotchie JM, Shin DS (2013) Deep brain stimulation of the substantia nigra pars reticulata improves forelimb akinesia in the hemiparkinsonian rat. J Neurophysiol 109:363–374PubMedCrossRef
Zurück zum Zitat Ten Kate M, Visser PJ, Bakardjian H, Barkhof F, Sikkes SAM, van der Flier WM, Scheltens P, Hampel H, Habert MO, Dubois B, Tijms BM (2018) Gray matter network disruptions and regional amyloid beta in cognitively normal adults. Front Aging Neurosci 10:67PubMedPubMedCentralCrossRef Ten Kate M, Visser PJ, Bakardjian H, Barkhof F, Sikkes SAM, van der Flier WM, Scheltens P, Hampel H, Habert MO, Dubois B, Tijms BM (2018) Gray matter network disruptions and regional amyloid beta in cognitively normal adults. Front Aging Neurosci 10:67PubMedPubMedCentralCrossRef
Zurück zum Zitat Tessa C, Giannelli M, Della Nave R, Lucetti C, Berti C, Ginestroni A, Bonuccelli U, Mascalchi M (2008) A whole-brain analysis in de novo Parkinson disease. AJNR Am J Neuroradiol 29:674–680PubMedPubMedCentralCrossRef Tessa C, Giannelli M, Della Nave R, Lucetti C, Berti C, Ginestroni A, Bonuccelli U, Mascalchi M (2008) A whole-brain analysis in de novo Parkinson disease. AJNR Am J Neuroradiol 29:674–680PubMedPubMedCentralCrossRef
Zurück zum Zitat Vaillancourt DE, Spraker MB, Prodoehl J, Abraham I, Corcos DM, Zhou XJ, Comella CL, Little DM (2009) High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology 72:1378–1384PubMedPubMedCentralCrossRef Vaillancourt DE, Spraker MB, Prodoehl J, Abraham I, Corcos DM, Zhou XJ, Comella CL, Little DM (2009) High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology 72:1378–1384PubMedPubMedCentralCrossRef
Zurück zum Zitat Valldeoriola F, Munoz E, Rumia J, Roldan P, Camara A, Compta Y, Marti MJ, Tolosa E (2019) Simultaneous low-frequency deep brain stimulation of the substantia nigra pars reticulata and high-frequency stimulation of the subthalamic nucleus to treat levodopa unresponsive freezing of gait in Parkinson’s disease: a pilot study. Parkinsonism Relat Disord 60:153–157PubMedCrossRef Valldeoriola F, Munoz E, Rumia J, Roldan P, Camara A, Compta Y, Marti MJ, Tolosa E (2019) Simultaneous low-frequency deep brain stimulation of the substantia nigra pars reticulata and high-frequency stimulation of the subthalamic nucleus to treat levodopa unresponsive freezing of gait in Parkinson’s disease: a pilot study. Parkinsonism Relat Disord 60:153–157PubMedCrossRef
Zurück zum Zitat van Rooden SM, Heiser WJ, Kok JN, Verbaan D, van Hilten JJ, Marinus J (2010) The identification of Parkinson’s disease subtypes using cluster analysis: a systematic review. Mov Disord 25:969–978PubMedCrossRef van Rooden SM, Heiser WJ, Kok JN, Verbaan D, van Hilten JJ, Marinus J (2010) The identification of Parkinson’s disease subtypes using cluster analysis: a systematic review. Mov Disord 25:969–978PubMedCrossRef
Zurück zum Zitat Vervoort G, Leunissen I, Firbank M, Heremans E, Nackaerts E, Vandenberghe W, Nieuwboer A (2016) Structural brain alterations in motor subtypes of Parkinson’s disease: evidence from probabilistic tractography and shape analysis. PLoS ONE 11:e0157743PubMedPubMedCentralCrossRef Vervoort G, Leunissen I, Firbank M, Heremans E, Nackaerts E, Vandenberghe W, Nieuwboer A (2016) Structural brain alterations in motor subtypes of Parkinson’s disease: evidence from probabilistic tractography and shape analysis. PLoS ONE 11:e0157743PubMedPubMedCentralCrossRef
Zurück zum Zitat Wang L, Yan Y, Zhang L, Liu Y, Luo R, Chang Y (2021) Substantia nigra neuromelanin magnetic resonance imaging in patients with different subtypes of Parkinson disease. J Neural Transm (vienna) 128:171–179CrossRef Wang L, Yan Y, Zhang L, Liu Y, Luo R, Chang Y (2021) Substantia nigra neuromelanin magnetic resonance imaging in patients with different subtypes of Parkinson disease. J Neural Transm (vienna) 128:171–179CrossRef
Zurück zum Zitat Willemsen MA, Verbeek MM, Kamsteeg EJ, de Rijk-van Andel JF, Aeby A, Blau N, Burlina A, Donati MA, Geurtz B, Grattan-Smith PJ, Haeussler M, Hoffmann GF, Jung H, de Klerk JB, van der Knaap MS, Kok F, Leuzzi V, de Lonlay P, Megarbane A, Monaghan H, Renier WO, Rondot P, Ryan MM, Seeger J, Smeitink JA, Steenbergen-Spanjers GC, Wassmer E, Weschke B, Wijburg FA, Wilcken B, Zafeiriou DI, Wevers RA (2010) Tyrosine hydroxylase deficiency: a treatable disorder of brain catecholamine biosynthesis. Brain 133:1810–1822PubMedCrossRef Willemsen MA, Verbeek MM, Kamsteeg EJ, de Rijk-van Andel JF, Aeby A, Blau N, Burlina A, Donati MA, Geurtz B, Grattan-Smith PJ, Haeussler M, Hoffmann GF, Jung H, de Klerk JB, van der Knaap MS, Kok F, Leuzzi V, de Lonlay P, Megarbane A, Monaghan H, Renier WO, Rondot P, Ryan MM, Seeger J, Smeitink JA, Steenbergen-Spanjers GC, Wassmer E, Weschke B, Wijburg FA, Wilcken B, Zafeiriou DI, Wevers RA (2010) Tyrosine hydroxylase deficiency: a treatable disorder of brain catecholamine biosynthesis. Brain 133:1810–1822PubMedCrossRef
Zurück zum Zitat Wu ST, Ma Y, Zhang K, Zhang JG (2012) Effect of deep brain stimulation on substantia nigra neurons in a rat model of Parkinson’s disease. Chin Med J (engl) 125:4072–4075 Wu ST, Ma Y, Zhang K, Zhang JG (2012) Effect of deep brain stimulation on substantia nigra neurons in a rat model of Parkinson’s disease. Chin Med J (engl) 125:4072–4075
Zurück zum Zitat Wu Y, Guo XY, Wei QQ, Ou RW, Song W, Cao B, Zhao B, Shang HF (2016) Non-motor symptoms and quality of life in tremor dominant vs postural instability gait disorder Parkinson’s disease patients. Acta Neurol Scand 133:330–337PubMedCrossRef Wu Y, Guo XY, Wei QQ, Ou RW, Song W, Cao B, Zhao B, Shang HF (2016) Non-motor symptoms and quality of life in tremor dominant vs postural instability gait disorder Parkinson’s disease patients. Acta Neurol Scand 133:330–337PubMedCrossRef
Zurück zum Zitat Xiang Y, Gong T, Wu J, Li J, Chen Y, Wang Y, Li S, Cong L, Lin Y, Han Y, Yin L, Wang G, Du Y (2017) Subtypes evaluation of motor dysfunction in Parkinson’s disease using neuromelanin-sensitive magnetic resonance imaging. Neurosci Lett 638:145–150PubMedCrossRef Xiang Y, Gong T, Wu J, Li J, Chen Y, Wang Y, Li S, Cong L, Lin Y, Han Y, Yin L, Wang G, Du Y (2017) Subtypes evaluation of motor dysfunction in Parkinson’s disease using neuromelanin-sensitive magnetic resonance imaging. Neurosci Lett 638:145–150PubMedCrossRef
Zurück zum Zitat Xu C, Zhuang P, Hallett M, Zhang Y, Li J, Li Y (2018) Parkinson’s disease motor subtypes show different responses to long-term subthalamic nucleus stimulation. Front Hum Neurosci 12:365PubMedPubMedCentralCrossRef Xu C, Zhuang P, Hallett M, Zhang Y, Li J, Li Y (2018) Parkinson’s disease motor subtypes show different responses to long-term subthalamic nucleus stimulation. Front Hum Neurosci 12:365PubMedPubMedCentralCrossRef
Zurück zum Zitat Zaidel A, Arkadir D, Israel Z, Bergman H (2009) Akineto-rigid vs. tremor syndromes in Parkinsonism. Curr Opin Neurol 22:387–393PubMedCrossRef Zaidel A, Arkadir D, Israel Z, Bergman H (2009) Akineto-rigid vs. tremor syndromes in Parkinsonism. Curr Opin Neurol 22:387–393PubMedCrossRef
Zurück zum Zitat Zhan W, Kang GA, Glass GA, Zhang Y, Shirley C, Millin R, Possin KL, Nezamzadeh M, Weiner MW, Marks WJ Jr, Schuff N (2012) Regional alterations of brain microstructure in Parkinson’s disease using diffusion tensor imaging. Mov Disord 27:90–97PubMedCrossRef Zhan W, Kang GA, Glass GA, Zhang Y, Shirley C, Millin R, Possin KL, Nezamzadeh M, Weiner MW, Marks WJ Jr, Schuff N (2012) Regional alterations of brain microstructure in Parkinson’s disease using diffusion tensor imaging. Mov Disord 27:90–97PubMedCrossRef
Zurück zum Zitat Zhang Y, Larcher KM, Misic B, Dagher A (2017) Anatomical and functional organization of the human substantia nigra and its connections. Elife 6 Zhang Y, Larcher KM, Misic B, Dagher A (2017) Anatomical and functional organization of the human substantia nigra and its connections. Elife 6
Metadaten
Titel
Nigral neuropathology of Parkinson’s motor subtypes coincide with circuitopathies: a scoping review
verfasst von
Jackson Tyler Boonstra
Hugo McGurran
Yasin Temel
Ali Jahanshahi
Publikationsdatum
19.07.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
Brain Structure and Function / Ausgabe 7/2022
Print ISSN: 1863-2653
Elektronische ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-022-02531-9

Weitere Artikel der Ausgabe 7/2022

Brain Structure and Function 7/2022 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Stuhltransfusion könnte Fortschreiten von Parkinson-Symptomen bremsen

03.05.2024 Parkinson-Krankheit Nachrichten

Kann eine frühzeitige Stuhltransplantation das Fortschreiten von Parkinson-Symptomen verlangsamen? Die Ergebnisse einer randomisierten Phase-2-Studie scheinen dafür zu sprechen.

Frühe Tranexamsäure-Therapie nützt wenig bei Hirnblutungen

02.05.2024 Hirnblutung Nachrichten

Erhalten Personen mit einer spontanen Hirnblutung innerhalb von zwei Stunden nach Symptombeginn eine Tranexamsäure-Therapie, kann dies weder die Hämatomexpansion eindämmen noch die Mortalität senken.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders scheint das auf weibliche Kranke zuzutreffen, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.