Look at the bars on the graph Which exposure methods appear to transmit Pd and cause and lesions

Structural and functional neuroimaging has long been used in the study of neurological disorders to elucidate disease mechanisms, to aid in diagnosis, and to assess potential new therapies. Clinical and research applications of functional neuroimaging to Parkinson’s disease (PD) and other parkinsonian disorders have advanced over the past decade, leading to novel diagnostic methods and contributing to the development of new therapies. This article will review how such imaging has been applied to the study of PD.

The defining pathologic characteristic of PD is the progressive loss of nigrostriatal dopaminergic function, leading to downstream changes in basal ganglia circuitry. Although direct imaging of the dopaminergic system with positron emission tomography (PET) or single-photon emission computed tomography (SPECT) can be valuable for assessing PD, there has also been great interest in understanding the functional consequences of dopaminergic pathology. Moreover, the symptoms of PD extend beyond the cardinal features of tremor, bradykinesia, and rigidity to other domains, including nonmotor symptoms. These manifestations of the disease cannot be attributed to simple dysfunction of the basal ganglia, but rather to widespread functional abnormalities involving a number of neuronal circuits (DeLong and Wichmann 2007). Accordingly, functional imaging with MRI (fMRI), PET, and SPECT has increasingly been used to assess neuronal connectivity, rather than focusing on single neurotransmitter systems. Moreover, functional imaging is not only able to study the processes underlying PD symptoms, but also can be applied to detecting subclinical and early disease and monitoring disease progression.

In this work we will briefly describe the principles behind PET, SPECT, and fMRI imaging and discuss the role of such imaging in assessing both motor and nonmotor features of PD. To date, the use of functional imaging in parkinsonian disorders has been largely restricted to research applications, which will be the focus of the article. We acknowledge that increasing efforts have been made to use these techniques in clinical practice (Poston 2011), but will not describe these in detail. Clinical applications include monitoring of preclinical disease (Tang et al. 2010a), studying at-risk populations (Piccini et al. 1999; Albin et al. 2000; Eisensehr et al. 2000; Ponsen et al. 2004; Sommer et al. 2004; Adams et al. 2005; Stiasny-Kolster et al. 2005), and using imaging as an adjunct method in diagnosis (Tang et al. 2010b). Last, functional imaging has been used in a number of clinical trials relating to PD (see Niethammer and Feigin 2011 for more information).

Dopaminergic imaging can be used to directly study the integrity of the nigrostriatal system in PD. [18F]-6-fluorodopa (FDOPA) PET measures the activity of AADC, thereby providing a means for quantifying the density of dopaminergic nerve terminals. Striatal FDOPA uptake is consistently and significantly reduced in patients with PD and other parkinsonian disorders (Brooks et al. 1990; Eidelberg et al. 1990; Vingerhoets et al. 1994; Thobois et al. 2004), and correlates with pathological measures (Snow et al. 1993). The reduction is not uniform throughout the striatum, with the smallest reduction in the caudate and an anterior–posterior gradient in the putamen (Morrish et al. 1996; Nurmi et al. 2001). Additionally, the reduction is asymmetrical, being more pronounced on the brain hemisphere contralateral to the more severe motor signs (Brooks et al. 1990; Eidelberg et al. 1990; Vingerhoets et al. 1997; Benamer et al. 2000; Pirker 2003; Filippi et al. 2005). Other radiotracers that provide measures of presynaptic dopaminergic function (e.g., DAT and vesicular monoamine transporter [VMAT] ligands), show similar findings, and all decline with disease progression (Vingerhoets et al. 1994; Hirano et al. 2010).

The degree of FDOPA reduction does not reflect all motor symptoms equally, but appears most strongly correlated with bradykinesia and, to a lesser extent, rigidity (Brooks et al. 1990; Eidelberg et al. 1990). Putaminal rather than caudate FDOPA uptake appears more closely related to motor function (Muller et al. 2000; Rinne et al. 2000; Ribeiro et al. 2002). Similarly, imaging of the dopamine transporter system (DAT) correlates with measures of bradykinesia, rigidity, gait, and facial expression (Pirker 2003). Interestingly, tremor, which is not uniformly responsive to dopaminergic therapy, does not appear to be correlated with dopaminergic deficits (Eidelberg et al. 1995a; Benamer et al. 2003; Pirker 2003; Isaias et al. 2007; Mure et al. 2011). Gait dysfunction also may not relate directly to dopaminergic loss. Bohnen et al. reported that cortical and thalamic cholinergic hypofunction rather than dopaminergic denervation is associated with fall risk in PD, perhaps relating to thalamic innervation from the pedunculopontine nucleus (PPN) (Bohnen et al. 2009). At least one study suggests that extrastriatal monoaminergic function as measured by FDOPA PET declines independently of the nigrostriatal system, although correlation of this decline with motor or nonmotor symptoms has not been established (Pavese et al. 2011).

FDG PET provides a means for mapping spatially distributed regional changes in cerebral glucose metabolism, and potentially relating these changes to specific diseases and symptoms. Spatial covariance analysis has been extensively used to detect network-level functional abnormalities in a variety of neurodegenerative disorders, including PD (Eidelberg et al. 1994; Eckert et al. 2007b), atypical parkinsonian syndromes such as multiple system atrophy (MSA) and progressive supranuclear palsy (PSP) (Eckert et al. 2008; Spetsieris et al. 2009), Huntington’s disease (Feigin et al. 2007), and dementia (Huang et al. 2007a; Habeck et al. 2008). The details of this method based on principal component analysis (PCA) have been reviewed elsewhere (Eidelberg 2009; Habeck and Stern 2010; Spetsieris and Eidelberg 2011). Briefly, the scaled subprofile model (SSM), a double-centered log-normalized PCA, is applied to multivoxel metabolic imaging data in a combined sample of scans from healthy subjects and patients. Once a pattern is identified that distinguishes one group from the other, its expression can be prospectively quantified on an individual basis (Spetsieris et al. 2006; Ma et al. 2007), and the resulting subject scores (i.e., the PCA scalars) can be correlated with clinical and physiological measures of interest.

Applying this method to resting-state FDG PET scans from PD patients has consistently revealed an abnormal disease-related spatial covariance pattern involving elements of the CSPTC circuitry (Eidelberg et al. 1994; Moeller et al. 1999; Eidelberg 2009). This Parkinson’s disease-related pattern (PDRP) is characterized by increased pallido-thalamic and pontine metabolic activity and relative reductions in premotor cortex, supplemental motor area, and parietal association regions (Fig. 1A). PDRP expression does not simply discriminate patients with PD from healthy subjects, but also distinguishes between PD and atypical parkinsonian syndromes (see below). The PDRP topography has been identified in several independent patient populations (Moeller et al. 1999; Eckert et al. 2007a). Subject scores denoting pattern expression in individual patients have been found to show excellent within-subject reproducibility on test–retest evaluation (Ma et al. 2007). Because metabolism and cerebral blood flow are coupled, at least in the absence of dopaminergic medication (Ma et al. 2007; Hirano et al. 2008), PDRP expression can also be measured in scans of resting cerebral perfusion obtained with [15O]H2O PET (Ma and Eidelberg 2007; Ma et al. 2007), 99mTc-ethylcysteinate dimer (ECD) SPECT (Feigin et al. 2002; Eckert et al. 2007b), or with arterial spin labeling MRI methods (Huang et al. 2007a; Melzer et al. 2011).

Look at the bars on the graph Which exposure methods appear to transmit Pd and cause and lesions

Abnormal metabolic networks in Parkinson’s disease. (A) Parkinson’s disease-related pattern (PDRP). This motor-related metabolic spatial covariance pattern is characterized by hypermetabolism in the thalamus, globus pallidus (GP), pons, and primary motor cortex, associated with relative metabolic reductions in the lateral premotor (PMC) and posterior parietal areas (Ma et al. 2007). (In the representative slices, relative metabolic increases are displayed in red; relative metabolic decreases are displayed in blue. Slices were overlaid on a standard MRI brain template.) (From Eidelberg 2009; reprinted, with permission, from Elsevier © 2009.) (B) PD tremor-related metabolic pattern (PDTP) identified using a within-subject network analysis of FDG PET scans from nine tremor-dominant PD patients scanned at baseline and during ventral-intermediate (Vim) thalamic deep brain stimulation (DBS) (Mure et al. 2011). This pattern is characterized by covarying increases in the metabolic activity of the sensorimotor cortex (SMC), cerebellum, pons, and the putamen. (From Mure et al. 2011; reprinted, with permission, from Elsevier © 2011.)

Individual subject PDRP expression values reproducibly correlate with Unified Parkinson’s Disease Rating Scale (UPDRS) motor ratings (Eidelberg et al. 1994, 1995b; Feigin et al. 2002; Lozza et al. 2004; Asanuma et al. 2006; Eidelberg 2009). Like dopaminergic imaging, abnormally elevated PDRP subject scores correlate mainly with bradykinesia and rigidity, rather than tremor (Antonini et al. 1998; Isaias et al. 2010; Mure et al. 2011). Moreover, these observations relate the abnormal PDRP functional topography to degeneration of nigrostriatal dopaminergic pathways (Eidelberg et al. 1990; Tang et al. 2010a). Of note, a similar pattern has recently been described in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) primate model of parkinsonian bradykinesia and rigidity (Peng et al. 2010; Ma et al. 2011).

PDRP expression increases continuously over time, in association with deterioration in motor function as well as putaminal dopaminergic function. The PDRP can be quantified separately in each hemisphere, and similar to dopaminergic imaging, PDRP expression is asymmetric, being worse on the side contralateral to the more affected body side (Eidelberg et al. 1995a; Tang et al. 2010a). Progression over time is similar in both hemispheres, preserving the initial asymmetry (Tang et al. 2010a). In addition, PDRP expression in individual subjects (i.e., the “subject scores”) has been found to correlate consistently with UPDRS motor ratings in multiple PD patient populations (Eidelberg et al. 1994, 1995b; Feigin et al. 2002; Lozza et al. 2004; Asanuma et al. 2006; see Eidelberg 2009). Nevertheless, correlations between longitudinal declines in presynaptic dopaminergic function, clinical deterioration, and increases in PDRP expression with advancing disease are modest (R2 ∼ 30%–40%), suggesting that these progression indices are not interchangeable (Eckert et al. 2007a; Huang et al. 2007a). Indeed, PDRP spatial topography conforms to the large-scale changes in regional synaptic activity predicted by experimental models of CSPTC circuitry (DeLong and Wichmann 2007). The biological relevance of PDRP is underscored by its relation to surgical treatments (Asanuma et al. 2006). For example, intraoperative recordings of subthalamic firing rates correlate closely with preoperative measurements of PDRP expression (Lin et al. 2008). Subthalamic lesioning and deep brain stimulation (DBS) both result in PDRP reductions of similar magnitude to those seen with levodopa treatment (Asanuma et al. 2006; Trost et al. 2006; Pourfar et al. 2009; Mattis et al. 2011).

Parkinsonian tremor has been linked to its own discrete spatial covariance pattern. A PD tremor-related pattern (PDTP) was identified using ordinal trends/cannonical variates analysis (OrT/CVA) (Habeck et al. 2005), a within-subject network mapping algorithm based on supervised (i.e., guided) PCA methods. FDG PET scans were acquired in tremor-predominant PD patients at baseline (tremor present) and during high-frequency stimulation (tremor suppressed) of the ventral-intermediate (Vim) thalamic nucleus (Mure et al. 2011) (Fig. 1B). This reproducible pattern is characterized by increased metabolic activity in the cerebellum and dorsal pons, primary motor cortex, and in the caudate and putamen. PDTP expression correlates with accelerometric tremor measurements and UPDRS tremor subscale ratings, but not with UPDRS akinesia-rigidity subscale ratings.

In recent years, resting-state imaging has been more extensively used in the investigation of brain disorders. Scanning PD patients at baseline, Helmich et al. (2010) found reduced functional connectivity in pathways linking the posterior putamen and in the inferior parietal cortex in PD patients, whereas connectivity between the anterior putamen and the pre-MSA, and between the caudate nucleus and the dorsal prefrontal cortex did not differ from normal. Other studies have not found reductions in functional connectivity in untreated PD. Rather, the data revealed normalization of baseline increases in cortico-striatal functional connectivity by dopaminergic treatment (Kwak et al. 2010). In addition to the pathways constituting the classical cortico-striato-pallido-thalamocortical (CSPTC) circuit model of motor pathways, recent attention has focused on the role the so-called “hyperdirect” pathway between motor cortex and STN plays in the motor physiology of PD (Brown et al. 2001; Priori et al. 2004; Bronte-Stewart et al. 2009). PD patients have been found to have increased resting-state functional connectivity between the STN and cortical motor regions (Baudrexel et al. 2011). Subgroup analysis revealed differences between PD patients with and without tremor: Compared to controls, tremor patients showed increased STN connectivity with the hand area of the primary motor cortex and the primary sensory cortex, whereas nontremor patients also had increased connectivity between the STN and midline cortical motor areas (Baudrexel et al. 2011). In this vein, tremor in PD has been related to transient increases in functional connectivity between the basal ganglia and motor cortex (Helmich et al. 2010). In PD patients with tremor, activity within the cerebellar pathways cofluctuates with tremor amplitude, whereas the globus pallidus (GPi and GPe), and putamen are transiently activated at the onset of tremor episodes. These data suggest that tremor in PD results from abnormal interactions between the basal ganglia and the cerebellothalamocortical circuit (Helmich et al. 2010).

Treatment of PD with levodopa is highly effective, but prolonged treatment will lead to motor and nonmotor complications in the majority of patients. Among these, levodopa-induced dyskinesias (LID) occur in up to 90% of PD patients after 9 yr of treatment (Ahlskog and Muenter 2001; Rajput et al. 2002). The risk of developing LID is associated with earlier age of onset, longer disease duration, as well as duration and dose of levodopa treatment (Fabbrini et al. 2007). A major feature of LID appears to be overactivity of the direct striatal pathway, but the pathophysiology of LID is complex, likely involving both pre- and postsynaptic alterations in the nigrostriatal system (Cenci and Lundblad 2006; Cenci and Lindgren 2007). Presynaptic dopaminergic denervation is presumed to be necessary for the development of LID, illustrated by the fact that healthy subjects with an intact presynaptic dopaminergic system do not develop LID (Constantinescu et al. 2007). Moreover, the development of LID presumably relates to an interaction between the endogenous dopaminergic denervation and application of levodopa, an exogenous dopamine precursor (Cenci and Lundblad 2006). Indeed, rodent and other animal models of LID have shown that abnormal presynaptic handling of levodopa as well as hyperactive postsynaptic dopamine responses play a role in LID (Cenci and Lindgren 2007). Pulsatile levodopa administration with rapid increase in synaptic dopamine concentrations appears to increase the risk of LID in animal models and PD patients (Smith et al. 2003; Pavese et al. 2006; Lindgren et al. 2010), and once established, there is a higher propensity for them to return even after prolonged interruption of treatment (Cenci and Lundblad 2006). Because peak-dose LID can be reliably triggered in many patients by levodopa administration, functional imaging has been able to add further insights into this complication of therapy.

On the presynaptic side, reduced FDOPA uptake (presumably reflecting greater disease severity) is associated with increased dyskinesia severity (Linazasoro et al. 2004). That study also suggested involvement of postsynaptic dopamine D2 receptor availability as measured with [11C]-raclopride PET (Linazasoro et al. 2004). Similarly, in a longitudinal study of levodopa versus ropinirole, patients treated with the agonist had less reduction in putaminal FDOPA uptake, with a lower incidence of LID (Whone et al. 2003). However, striatal dopaminergic denervation is not sufficient for the development of LID. Comparing patients with LID on low-dose levodopa with patients without LID on high-dose levodopa, Linazasoro et al. (2009) found equally reduced striatal DAT binding. Dopamine turnover may play a role in this regard. Early in the disease, synaptic dopamine turnover has been found to be increased when estimated by kinetic modeling of FDOPA time-activity curves (TACs) over prolonged scanning epochs (Sossi et al. 2002, 2004). The relative increase in dopamine turnover observed in early-stage disease was even more pronounced in younger patients, possibly contributing to larger swings in dopamine levels, which explains the greater susceptibility to motor complications seen in those patients (Sossi et al. 2006). Higher synaptic dopamine levels in patients with LID were also shown using [11C]-raclopride PET, and the size of the levodopa-mediated change in radioligand binding affinity correlates with dyskinesia severity (de la Fuente-Fernandez et al. 2004; Pavese et al. 2006). Of note, a multitracer study of VMAT and DAT binding revealed relative down-regulation of dopamine reuptake compared to nerve terminal loss (Troiano et al. 2009). This adaptation may help with symptoms early in the disease course, but can give rise to oscillatory synaptic dopamine and concomitant motor complications as the disease progresses. Animal studies have suggested an increase of D1 receptor numbers in response to long-term levodopa treatment (Rioux et al. 1997). However, consistent changes in striatal D1 and D2 neuroreceptor binding have not been shown in human PD patients (Turjanski et al. 1997; Dentresangle et al. 1999).

Other neurotransmitter systems have been implicated in the development of LID based on animal or postmortem studies. These include the opioid system, which acts as a modulator of dopamine and a cotransmitter within GABAergic neurons (Fox et al. 2006; Hallett and Brotchie 2007), the adenosine system (Calon et al. 2004), the serotonergic system (Carta et al. 2007; Carlsson et al. 2009), and the α2 adrenergic system (Fox et al. 2001; Rascol et al. 2001). Piccini et al. (1997) found reduced binding of [11C]diphrenorphine (a nonselective marker of opioid receptors) in the striatum, thalamus, and anterior cingulate of PD patients with LID compared to without. LID severity was associated with reduced putaminal binding, possibly indicating increased opioid receptor occupancy by endogenous ligands in patients with LID.

Both dopamine agonists and levodopa trigger localized increases in cerebral blood flow (Tuor et al. 1986; Beck et al. 1988). Functional imaging studies utilizing Xenon-133 have found similar increases in PD patients and normal subjects in response to dopaminergic treatment (Montastruc et al. 1987; Kobari et al. 1995). Increased blood flow in the striatum, globus pallidus, and thalamus has been associated with adverse reactions to levodopa in PD patients, whereas those without adverse reactions show decreased blood flow (Henriksen and Boas 1985). In a motor activation study, patients with LID showed significant overactivation in the SMA and bilateral primary motor cortex measured with Xenon-133 compared to those without LID (Rascol et al. 1998). H215O PET has also been used to evaluate changes in blood flow after levodopa administration. Hershey et al. (1998) found that patients with LID showed increased perfusion in the ventrolateral thalamus even at levodopa doses insufficient to trigger LID (Hershey et al. 1998). In general, the blood flow changes have been attributed to direct effects of dopamine on local microvasculature (Iadecola 1998; Edvinsson and Krause 2002).

The changes in blood flow in response to levodopa contrast with changes in regional metabolism. Untreated PD patients show increased glucose metabolism in the putamen, globus pallidus, ventral thalamus, and dorsal pons (Fukuda et al. 2001a,b; Huang et al. 2007b). These regional abnormalities have been associated with disease-related changes in local synaptic activity at salient nodes of the PDRP metabolic network (Eidelberg 2009). In the rest state, tight correlations have been shown between regional cerebral metabolic rate (CMR) and blood flow (CBF), consistent with the notion that both measures reflect local synaptic activity (Ma and Eidelberg 2007). Thus, PDRP expression measured from FDG PET scans, H2O PET scans, and perfusion-weighted MRI scans in the same subjects are significantly correlated (Ma et al. 2007, 2010). However, dopaminergic treatment has been found to dissociate (“uncouple”) these effects (Hirano et al. 2008). Effective treatment with levodopa reduces abnormally elevated metabolic activity (Feigin et al. 2001; Asanuma et al. 2006), but increases basal ganglia CBF (Kobari et al. 1995; Hershey et al. 2003). Indeed, PDRP network modulation is dissociated between CBF and CMR in PD patients undergoing levodopa treatment but not during STN stimulation of comparable clinical efficacy. The phenomenon of flow-metabolism dissociation observed at the network level was localized regionally to the putamen and dorsal pons. In these areas, the increases in rCBF with levodopa treatment are greater in patients with LID, compatible with an exaggeration of microvascular responsiveness to monoamines in these individuals (Hirano et al. 2008). In animal models, levodopa treatment induces endothelial proliferation and angiogenesis, which may provide a possible basis for this observation (Westin et al. 2006; Lindgren et al. 2010).

The clinical diagnosis and definition of PD is based on motor signs and symptoms. Nevertheless, nonmotor symptoms may be prominent, and can precede motor symptoms (Tolosa et al. 2009; Lang 2011), and they can encompass a variety of categories, such as neuropsychiatric, sleep, or autonomic dysfunction (Lim and Lang 2010). Among the premotor symptoms, hyposmia and idiopathic rapid eye movement (REM) sleep behavior disorder both confer a higher risk of developing PD, and dopaminergic imaging may be abnormal in such patients as will be discussed further below. In this section we will focus on imaging applications relating to cognitive dysfunction.

Cognitive functioning can be substantially impaired in PD, playing a major role in clinical disability and in overall treatment outcome. The point prevalence of dementia in PD may be as high as 31%, with substantially higher cumulative prevalence (Aarsland et al. 2005; Aarsland and Kurz 2010). Even without dementia, cognitive difficulties can be present in early PD, with features similar but less severe to those seen in PD patients with dementia (PDD) (Green et al. 2002; Huang et al. 2007b).

Functional imaging studies have implicated multiple mechanisms for cognitive dysfunction and dementia in PD, including dopaminergic depletion, cholinergic dysfunction, as well as protein aggregation and cortical pathology (i.e., Lewy body formation). As with motor function, functional imaging in the resting state as well as activation paradigms has been applied to the study of cognitive dysfunction in PD.

FDG PET has consistently shown cortical hypometabolism in PD. Even patients without apparent cognitive impairment show hypometabolism in the frontal and occipital cortices (Hosokai et al. 2009). This hypometabolism appears more widespread in PD patients with mild cognitive impairment (MCI), encompassing prefrontal and lateral parietal cortices (Mentis et al. 2002; Huang et al. 2007a, 2008) and may include occipital and medial frontal regions (Hosokai et al. 2009; Borghammer et al. 2010). Indeed, comparisons of patients with and without MCI, or patients with single- and multiple-domain MCI suggest that the degree and topography of hypometabolism reflects the extent of cognitive dysfunction (Peppard et al. 1990; Yong et al. 2007; Huang et al. 2008; Hosokai et al. 2009).

Early studies using PET imaging and network analysis suggested separate patterns of metabolic dysfunction relating to cognitive, affective, and motor function (Mentis et al. 2002; Carbon et al. 2003; Lozza et al. 2004). Applying spatial covariance analysis to FDG PET data from a cohort of nondemented PD patients, Huang et al. (2007a) identified a significant covariance pattern that correlated with cognitive performance, particularly involving executive functioning. This pattern, termed the PD-related cognitive pattern (PDCP), is characterized by metabolic reductions in frontal and parietal association areas, and increases in the cerebellar vermis and dentate nuclei (Fig. 2A) (Huang et al. 2007a). PDCP expression increased with worsening of cognitive impairment (see Fig. 2B,C) (Huang et al. 2008; Eidelberg 2009). The PD-related metabolic patterns are functionally independent. Although PDCP and PDRP expression progress over time, only the changes in PDRP expression correlate with concurrent declines in striatal dopaminergic function (Huang et al. 2007b). On group analysis, PDCP expression, unlike PDRP, does not respond to treatment of parkinsonian motor signs with levodopa or DBS (Huang et al. 2007a). Nevertheless, levodopa does affect cognitive function in some patients, and levodopa-mediated changes in verbal learning correlate with baseline PDCP expression. Moreover, in patients with meaningful improvement on levodopa there is a concurrent reduction in PDCP expression (Mattis et al. 2011).

Look at the bars on the graph Which exposure methods appear to transmit Pd and cause and lesions

Parkinson’s disease-related cognitive pattern: validation and correlates. (A) Parkinson’s disease-related cognitive pattern (PDCP). This cognition-related metabolic spatial covariance pattern is characterized by hypometabolism of dorsolateral prefrontal cortex (PMC), rostral supplementary motor area (preSMA), precuneus, and posterior parietal regions, associated with relative metabolic increases in the cerebellum (Huang et al. 2007a). (In the representative slices, relative metabolic increases are displayed in red; relative metabolic decreases are displayed in blue. Slices were overlaid on a standard MRI brain template.) (B) PDCP expression correlates with performance on neuropsychological tests of memory and executive functioning in nondemented PD patients. For the California Verbal Learning Test: Sum 1 to 5 (CVLT sum), this correlation was significant for the entire cohort (n = 56: r = −0.67, p < 0.001), as well as for the original group used for pattern derivation (circles, n = 15: r = −0.71, p = 0.003) and in two prospective validation groups (squares, n = 25: r = −0.53, p = 0.007; triangles, n = 16: r = −0.80, p < 0.001) (Huang et al. 2007a). (C) Bar graph of PDCP expression (mean ± SE) in PD patients with dementia (PDD), multiple-domain mild cognitive impairment (MCI[m]), single-domain mild cognitive impairment (MCI([s]), PD patients without mild cognitive impairment (MCI[−]), and in healthy control subjects. There was a significant difference in PDCP expression across the patient and control groups (F(4,70) = 8.87, p < 0.001; one-way ANOVA) and among the PD groups (F(3,56) = 4.84; p < 0.005), with higher expression in the PDD and MCI(m) cohorts compared to the MCI(−) cohort (p < 0.03; Tukey-Kramer HSD). For each PD group, PDCP expression was separately compared to healthy control values using Student's t-tests. The asterisks denote significant increases in network activity relative to controls (*p < 0.05, **p < 0.005, ***p < 0.0001) in all PD categories including MCI(−). (From Eidelberg 2009; reprinted, with permission, from Elsevier © 2009.)

Both beneficial and detrimental effects of PD treatment on cognition might be influenced by dopamine levels in different parts of the striatum (which in turn are affected by disease severity, genetic factors, and treatment) (Cools 2006; Mattis et al. 2011). For example, in a series of studies of motor sequence learning, GPi and STN stimulation were associated with improved performance, whereas levodopa was not (Fukuda et al. 2002; Carbon et al. 2003; Feigin et al. 2003; Argyelan et al. 2008). However, patients with poor baseline performance actually benefited from levodopa whereas patients with good baseline performance declined (Fig. 3) (Argyelan et al. 2008). Specifically, levodopa suppressed the normal deactivation in the ventromedial prefrontal cortex (vmPFC) that occurs during motor sequence learning. In good learners, deactivation of vmPFC was normal, but became abnormally depressed with levodopa. Other studies with fMRI or H215O PET have also found levodopa-mediated normalization of prefrontal activations, suggesting potential beneficial effects of levodopa on mesocortical pathways (Cools et al. 2002; Mattay et al. 2002).

Look at the bars on the graph Which exposure methods appear to transmit Pd and cause and lesions

Learning-related deactivation responses in the ventromedial prefrontal cortex (vmPFC) displayed as an inverted-U function. In addition to the influence of baseline phenotype on the cognitive response to dopaminergic therapy (Argyelan et al. 2008), we also found a significant influence of genotype. The observation that the effect of treatment on the vmPFC deactivation response varied according to COMT val158met genotype suggests that this physiologic effect is linked to intrinsic differences in prefrontal dopamine pools. Specifically, in carriers of valine alleles (VAL, i.e., valine homozygotes and val/met heterozygotes), levodopa served to suppress learning-related deactivation in this region. In contrast, recovery of this response tended to occur in individuals without this allele (MET, i.e., methionine homozygotes). The individual data were found to conform to an inverted-U function. Bands of low and high dopamine at the edges of the curve (shaded areas) represent zones of optimal function in which local deactivation responses occur during task performance. (From Argyelan et al. 2008; reprinted, with permission, from the Society for Neuroscience © 2008.)

Patients with PD commonly show impairments in executive tasks (van Eimeren et al. 2009). During the past decade there has been recognition that performance of many executive or attentional tasks may require deactivation of a baseline network. This network, termed the default mode network (DMN), identified with fMRI or PET imaging, includes the medial prefrontal cortex, posterior cingulate cortex, precuneus, and lateral parietal and temporal cortices (Raichle et al. 2001; Raichle and Snyder 2007). Impaired deactivation of the DMN may be seen in normal aging as well as in patients with dementia or neuropsychiatric illnesses (Lustig et al. 2003; Grady et al. 2006; Calhoun et al. 2008). Given that the loss of dopamine in PD leads to functional impairment of CSPTC pathways and modulation of frontal lobe activity, it is not surprising that deactivation of the DMN would be impaired in this disease.

To date, few studies have addressed the DMN in PD. Using fMRI and a semantic event sequencing task, Tinaz et al. found that in contrast to controls, patients with PD did not show decreased activation of the DMN during task performance (Tinaz et al. 2008). Similarly, during a Montreal card sorting task, PD patients did not only show less deactivation of the parietal DMN nodes, but even showed decreased activation of these same areas during the control task (van Eimeren et al. 2009). This reverse pattern of activation and deactivation differs from that seen in normal aging (Lustig et al. 2003) and may suggest PD-specific abnormalities relating to dopaminergic dysfunction. In healthy young subjects the complexity of a task correlates with the degree of DMN deactivation, a relationship that may be lost in healthy elderly subjects and PD patients (Nagano-Saito et al. 2009). Interestingly, in PD patients and healthy controls, apomorphine seems to restore this correlation (Nagano-Saito et al. 2009).

Reduced striatal dopaminergic function has been observed in PD patients with and without cognitive dysfunction (Ito et al. 2002; O’Brien et al. 2004; Bruck et al. 2005; Hilker et al. 2005). That said, dopaminergic dysfunction in the caudate may contribute to the cognitive deficits of PD. Indeed, PD patients with dementia have been shown to have greater loss of tracer uptake in the caudate than those without dementia (Ito et al. 2002; O’Brien et al. 2004). Jokinen et al. (2009) found a significant correlation between verbal and visual memory functioning and caudate FDOPA uptake, although they also noted additional associations with hippocampal and prefrontal atrophy. Several other studies have shown relationships between reductions in caudate dopaminergic tracer uptake and impaired performance on tests of memory, executive, and frontal lobe function (Marie et al. 1999; Muller et al. 2000; Rinne et al. 2000; Bruck et al. 2001; van Beilen et al. 2008). During sequence learning, PD patients showed a correlation between correct acquisition of targets and caudate DAT binding (Carbon et al. 2004). Moreover, normal subjects showed close correlations between caudate DAT binding and learning-related activation in the dorsolateral and ventral prefrontal cortices, a relationship that was lost in PD patients (Carbon et al. 2004). Indeed, we have recently found that PDCP expression correlates with caudate but not putamen DAT binding (Tang et al. 2011).

Extrastriatal dopaminergic function may also be linked to cognitive performance. For example, FDOPA uptake in the dorsolateral frontal cortex may be increased in drug-naïve patients and is associated with worse performance on tests of sustained attention (Bruck et al. 2005). Conversely, increased FDOPA uptake in the medial frontal cortex and anterior cingulate is associated with improved performance on tests of suppressed attention. Although this may reflect compensatory changes of dopaminergic transmission in early PD, the same findings could also reflect increased AADC activity relating to other neurotransmitter systems (serotonergic and noradrenergic) in those regions (Rakshi et al. 1999; Bruck et al. 2005).

Postmortem examinations in patients with PD have consistently reported evidence of cholinergic dysfunction in thalamus, striatum, and cortical areas (Arendt et al. 1983; Whitehouse et al. 1983; 1988; Aubert et al. 1992; Pimlott et al. 2004; Schmaljohann et al. 2006), and Ruberg et al. (1986) reported greater reductions in the frontal cortex of demented PD patients compared to their counterparts. Unlike Alzheimer’s disease (AD), there appears to be a loss of cholinergic function in the brain stem in PD (Jellinger 1988; Gai et al. 1991). Various radiotracers can be used for in vivo imaging of acetylcholinesterase (AChE), nicotinic and muscarinic acetylcholine receptors (nACHR and mAChR), and acetylcholine vesicular transporter (VAChT). Both [11C]-methyl-4-piperidinly proprionate (PMP) and [11C]-methyl-4-piperidyl acetate (MP4A) can be used to assess AChE function (Irie et al. 1996). Cortical AChE activity is reduced in patients with PD, PDD, and diffuse Lewy body disease (DLB). This reduction is more severe in PD patients with dementia (ranging from 17% to 29.7%) than in their nondemented counterparts (10.2%–12.9%) (Shinotoh et al. 1999; Bohnen et al. 2003; 2009; Shimada et al. 2009). Moreover, cortical AChE deficits may be larger in PDD and DLB compared to AD (Herholz et al. 2000; Bohnen et al. 2003). This decline in cortical cholinergic function appears to begin early in the disease process, and worsens with the onset of dementia (Shimada et al. 2009). Cholinergic dysfunction is less correlated with disease duration and severity of motor symptoms than with tests of attention and cognitive function (Bohnen et al. 2006). Similarly, PD and PDD patients show reductions in striatal FDOPA uptake, although PDD patients have comparatively lower cortical MP4A binding (Hilker et al. 2005). Nevertheless, in PDD, reductions in cortical MP4A and striatal FDOPA binding are closely linked, suggesting that combined decline in the two neurotransmitter systems plays a role in these patients (Hilker et al. 2005).

Imaging of the VAChT system with [123I]-iodobenzovesamicol (IBVM) has similarly revealed cortical reductions in nondemented PD patients, predominantly in the parietal and occipital cortices, with more widespread and severe reductions in PDD (Kuhl et al. 1996). Studies of nAChR in PD have shown a consistent reduction in subcortical tracer binding, with most also reporting reduced cortical receptors (Fujita et al. 2006; Oishi et al. 2007; Kas et al. 2009; Meyer et al. 2009). Although these studies were performed largely in nondemented PD patients, O’Brien et al. (2008) used [123I]-5-I-A-85380 SPECT to evaluate nAChR activity in patients with DLB. They found reduced tracer binding in the striatum, frontal, temporal, and cingulate areas, with a relative increase in the occipital cortex, perhaps suggesting a link between cholinergic function and visual hallucinations in that area (O’Brien et al. 2008). In contrast to the nicotinic system, imaging studies of mAChR have reported increased frontal and occipital receptor activity in patients with PD and PDD (Asahina et al. 1995, 1998; Colloby et al. 2006). However, demented patients with PSP did not show this increase, perhaps implying that the muscarinergic system is not directly linked to cognitive dysfunction (Asahina et al. 1998). Possibly the increase in occipital mAChR activity, like the increase in occipital nAChR activity, underlies the visual disturbances seen in DLB and PDD (Colloby et al. 2006). Overall, cortical cholinergic dysfunction appears to be a feature of cognitive impairment in PD. These observations are consistent with the observed improvement in cognition in clinical trials of AchE inhibitors in demented PD patients (Giladi et al. 2003; Emre et al. 2004).

As in other neurodegenerative diseases, the pathology of PD is associated with the accumulation of misfolded proteins, namely, Lewy bodies (Galvin et al. 2001). Lewy bodies, neuronal intracellular inclusions that contain α-synuclein, are more widespread in PDD and DLB. Additionally, β-amyloid deposition, i.e., Alzheimer pathology, is seen in both PDD and DLB, and may be associated with poor clinical outcome (Jellinger et al. 2002). The past few years have seen the development of radioligands for in vivo imaging of amyloid deposition. Of these, 2-(1-{6-[(2-[18F]Fluoroethyl)(methyl)amino]-2-naphthy}ethylidene)malononitrile (FDDNP) is relatively nonspecific, labeling senile plaques and neurofibrillary tangles in AD, prion plaques, amyloid deposits, and cortical Lewy bodies in vitro (Agdeppa et al. 2003; Smid et al. 2006). In contrast, [N-methyl-C-11]2-(4′-methylaminophenyl)-6-hydroxybenzothiazole (Klunk et al. 2004) (Pittsburgh Compound B, PIB) binds strongly to amyloid plaques in AD, but not significantly to Lewy bodies (Fodero-Tavoletti et al. 2007; Ye et al. 2008). Indeed, limited autopsy studies suggest that in PDD or DLB, in vivo PIB imaging does reflect β-amyloid rather than Lewy body pathology (Bacskai et al. 2007; Burack et al. 2010). For instance, Burack et al. (2010) reported on three PDD cases that had PIB imaging within 15 months of their death. Two of the three had elevated PIB uptake. All three had abundant cortical Lewy bodies, but the two individuals with elevated PIB uptake were also found to have abundant diffuse Aβ plaques.

Several studies have used PIB to evaluate patients with synucleinopathies (PD, PDD, DLB) (Brooks 2009). Generally speaking, cognitively normal PD patients do not have elevated PIB binding (Edison et al. 2008; Gomperts et al. 2008; Johansson et al. 2008; Maetzler et al. 2009). In contrast, patients with DLB show abnormally elevated cortical PIB binding, at levels comparable to patients with AD (Gomperts et al. 2008; Maetzler et al. 2009). PDD, which may be part of the same spectrum as DLB, or a separate disease more closely related to PD, is generally associated with normal cortical PIB binding. When PIB is elevated in PDD, it is generally lower than in DLB patients (Fig. 4A) (Edison et al. 2008; Gomperts et al. 2008; Brooks 2009), and amyloid load may not correlate with measures of dementia severity (Gomperts et al. 2008). Nevertheless, in the synucleinopathies, occipital PIB binding was higher compared to that observed in AD patients, and amyloid deposition in the parietal and posterior cingulate region was related to visuospatial impairment (Gomperts et al. 2008). In another recent study amyloid deposition was associated with lower MMSE scores in patients with dementia (DLB and PDD), but a majority of demented patients did not have elevated PIB binding (Maetzler et al. 2009), suggesting that other factors play a role in cognitive decline in Lewy body disease.

Look at the bars on the graph Which exposure methods appear to transmit Pd and cause and lesions

Protein aggregation in Parkinson’s disease. (A) 11C-PIB uptake in a healthy volunteer, PDD subject without significant amyloid, and two DLB patients with a significant amyloid load. (From Brooks 2009; reprinted, with permission, from John Wiley & Sons © 2009.) (B) Distribution of cortical protein aggregation in regions of interest (ROIs) computed using a standard brain atlas. Binding values for [11C]-PIB and [18F]-FDDNP were estimated from PET images acquired in PD patients by measuring the target-to-cerebellum ratio >60–90 min following radiotracer injection. The values presented on the Y axis represent the difference in uptake of the two tracers in regions with metabolic reduction in the same subject.

Although FDDNP and PIB imaging in combination might be useful for in vivo measurements of Lewy body pathology via a subtraction approach, to date there are no published studies formally evaluating this approach in PD. We have studied a 64-yr-old PD patient with FDG, PIB, and FDDNP PET imaging. This individual had elevated PDCP expression measured with FDG PET. We next used an automated region-of-interest approach to assess specific PIB and FDDNP binding relative to the cerebellum. In the majority of the regions, PIB and FDDNP uptake were closely matched, but in four areas there was significant discrepancy (Fig. 4B). For instance, in the anterior cingulate region, FDDNP binding was elevated whereas PIB binding was not. Furthermore, metabolic rates were reduced in areas of increased protein aggregation, suggesting that key nodes of the PDCP network show a relationship between reduced glucose utilization and increased protein aggregation. However, it remains to be seen if PIB and FDDNP indeed can reflect regional differences in pathology in a large cohort of PD patients.

  • Aarsland D, Kurz MW 2010. The epidemiology of dementia associated with Parkinson disease. J Neurol Sci 289: 18–22 [PubMed] [Google Scholar]
  • Aarsland D, Zaccai J, Brayne C 2005. A systematic review of prevalence studies of dementia in Parkinson’s disease. Mov Disord 20: 1255–1263 [PubMed] [Google Scholar]
  • Achard S, Salvador R, Whitcher B, Suckling J, Bullmore E 2006. A resilient, low-frequency, small-world human brain functional network with highly connected association cortical hubs. J Neurosci 26: 63–72 [PMC free article] [PubMed] [Google Scholar]
  • Adams JR, van Netten H, Schulzer M, Mak E, McKenzie J, Strongosky A, Sossi V, Ruth TJ, Lee CS, Farrer M, et al. 2005. PET in LRRK2 mutations: Comparison to sporadic Parkinson’s disease and evidence for presymptomatic compensation. Brain 128: 2777–2785 [PubMed] [Google Scholar]
  • Agdeppa ED, Kepe V, Liu J, Small GW, Huang SC, Petric A, Satyamurthy N, Barrio JR 2003. 2-Dialkylamino-6-acylmalononitrile substituted naphthalenes (DDNP analogs): Novel diagnostic and therapeutic tools in Alzheimer’s disease. Mol Imaging Biol 5: 404–417 [PubMed] [Google Scholar]
  • Ahlskog JE, Muenter MD 2001. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord 16: 448–458 [PubMed] [Google Scholar]
  • Albin RL, Koeppe RA, Chervin RD, Consens FB, Wernette K, Frey KA, Aldrich MS 2000. Decreased striatal dopaminergic innervation in REM sleep behavior disorder. Neurology 55: 1410–1412 [PubMed] [Google Scholar]
  • Antonini A, Moeller JR, Nakamura T, Spetsieris P, Dhawan V, Eidelberg D 1998. The metabolic anatomy of tremor in Parkinson’s disease. Neurology 51: 803–810 [PubMed] [Google Scholar]
  • Arendt T, Bigl V, Arendt A, Tennstedt A 1983. Loss of neurons in the nucleus basalis of Meynert in Alzheimer’s disease, paralysis agitans and Korsakoff’s disease. Acta Neuropathol 61: 101–108 [PubMed] [Google Scholar]
  • Argyelan M, Carbon M, Ghilardi MF, Feigin A, Mattis P, Tang C, Dhawan V, Eidelberg D 2008. Dopaminergic suppression of brain deactivation responses during sequence learning. J Neurosci 28: 10687–10695 [PMC free article] [PubMed] [Google Scholar]
  • Asahina M, Shinotoh H, Hirayama K, Suhara T, Shishido F, Inoue O, Tateno Y 1995. Hypersensitivity of cortical muscarinic receptors in Parkinson’s disease demonstrated by PET. Acta Neurol Scand 91: 437–443 [PubMed] [Google Scholar]
  • Asahina M, Suhara T, Shinotoh H, Inoue O, Suzuki K, Hattori T 1998. Brain muscarinic receptors in progressive supranuclear palsy and Parkinson’s disease: A positron emission tomographic study. J Neurol Neurosurg Psychiatry 65: 155–163 [PMC free article] [PubMed] [Google Scholar]
  • Asanuma K, Tang C, Ma Y, Dhawan V, Mattis P, Edwards C, Kaplitt MG, Feigin A, Eidelberg D 2006. Network modulation in the treatment of Parkinson’s disease. Brain 129: 2667–2678 [PMC free article] [PubMed] [Google Scholar]
  • Aubert I, Araujo DM, Cecyre D, Robitaille Y, Gauthier S, Quirion R 1992. Comparative alterations of nicotinic and muscarinic binding sites in Alzheimer’s and Parkinson’s diseases. J Neurochem 58: 529–541 [PubMed] [Google Scholar]
  • Bacskai BJ, Frosch MP, Freeman SH, Raymond SB, Augustinack JC, Johnson KA, Irizarry MC, Klunk WE, Mathis CA, Dekosky ST, et al. 2007. Molecular imaging with Pittsburgh Compound B confirmed at autopsy: A case report. Arch Neurol 64: 431–434 [PubMed] [Google Scholar]
  • Baudrexel S, Witte T, Seifried C, von Wegner F, Beissner F, Klein JC, Steinmetz H, Deichmann R, Roeper J, Hilker R 2011. Resting state fMRI reveals increased subthalamic nucleus-motor cortex connectivity in Parkinson’s disease. Neuroimage 55: 1728–1738 [PubMed] [Google Scholar]
  • Beck T, Vogg P, Krieglstein J 1988. Uncoupling of cerebral blood flow and glucose utilization by dihydroergocristine in the conscious rat. N-S Arch Pharmacol 338: 82–87 [PubMed] [Google Scholar]
  • Benamer HT, Patterson J, Wyper DJ, Hadley DM, Macphee GJ, Grosset DG 2000. Correlation of Parkinson’s disease severity and duration with 123I-FP-CIT SPECT striatal uptake. Mov Disord 15: 692–698 [PubMed] [Google Scholar]
  • Benamer HT, Oertel WH, Patterson J, Hadley DM, Pogarell O, Hoffken H, Gerstner A, Grosset DG 2003. Prospective study of presynaptic dopaminergic imaging in patients with mild parkinsonism and tremor disorders: Part 1. Baseline and 3-month observations. Mov Disord 18: 977–984 [PubMed] [Google Scholar]
  • Bohnen NI, Kaufer DI, Ivanco LS, Lopresti B, Koeppe RA, Davis JG, Mathis CA, Moore RY, DeKosky ST 2003. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: An in vivo positron emission tomographic study. Arch Neurol 60: 1745–1748 [PubMed] [Google Scholar]
  • Bohnen NI, Kaufer DI, Hendrickson R, Ivanco LS, Lopresti BJ, Constantine GM, Mathis Ch A, Davis JG, Moore RY, Dekosky ST 2006. Cognitive correlates of cortical cholinergic denervation in Parkinson’s disease and parkinsonian dementia. J Neurol 253: 242–247 [PubMed] [Google Scholar]
  • Bohnen NI, Muller ML, Koeppe RA, Studenski SA, Kilbourn MA, Frey KA, Albin RL 2009. History of falls in Parkinson disease is associated with reduced cholinergic activity. Neurology 73: 1670–1676 [PMC free article] [PubMed] [Google Scholar]
  • Borghammer P, Chakravarty M, Jonsdottir KY, Sato N, Matsuda H, Ito K, Arahata Y, Kato T, Gjedde A 2010. Cortical hypometabolism and hypoperfusion in Parkinson’s disease is extensive: Probably even at early disease stages. Brain Struct Funct 214: 303–317 [PubMed] [Google Scholar]
  • Bronte-Stewart H, Barberini C, Koop MM, Hill BC, Henderson JM, Wingeier B 2009. The STN β-band profile in Parkinson’s disease is stationary and shows prolonged attenuation after deep brain stimulation. Exp Neurol 215: 20–28 [PubMed] [Google Scholar]
  • Brooks DJ 2005. Positron emission tomography and single-photon emission computed tomography in central nervous system drug development. NeuroRx 2: 226–236 [PMC free article] [PubMed] [Google Scholar]
  • Brooks DJ 2009. Imaging amyloid in Parkinson’s disease dementia and dementia with Lewy bodies with positron emission tomography. Mov Disord 24 Suppl 2: S742–S747 [PubMed] [Google Scholar]
  • Brooks DJ, Ibanez V, Sawle GV, Quinn N, Lees AJ, Mathias CJ, Bannister R, Marsden CD, Frackowiak RS 1990. Differing patterns of striatal 18F-dopa uptake in Parkinson’s disease, multiple system atrophy, and progressive supranuclear palsy. Ann Neurol 28: 547–555 [PubMed] [Google Scholar]
  • Brown P, Oliviero A, Mazzone P, Insola A, Tonali P, Di Lazzaro V 2001. Dopamine dependency of oscillations between subthalamic nucleus and pallidum in Parkinson’s disease. J Neurosci 21: 1033–1038 [PMC free article] [PubMed] [Google Scholar]
  • Bruck A, Portin R, Lindell A, Laihinen A, Bergman J, Haaparanta M, Solin O, Rinne JO 2001. Positron emission tomography shows that impaired frontal lobe functioning in Parkinson’s disease is related to dopaminergic hypofunction in the caudate nucleus. Neurosci Lett 311: 81–84 [PubMed] [Google Scholar]
  • Bruck A, Aalto S, Nurmi E, Bergman J, Rinne JO 2005. Cortical 6-[18F]fluoro-L-dopa uptake and frontal cognitive functions in early Parkinson’s disease. Neurobiol Aging 26: 891–898 [PubMed] [Google Scholar]
  • Burack MA, Hartlein J, Flores HP, Taylor-Reinwald L, Perlmutter JS, Cairns NJ 2010. In vivo amyloid imaging in autopsy-confirmed Parkinson disease with dementia. Neurology 74: 77–84 [PMC free article] [PubMed] [Google Scholar]
  • Calhoun VD, Maciejewski PK, Pearlson GD, Kiehl KA 2008. Temporal lobe and “default” hemodynamic brain modes discriminate between schizophrenia and bipolar disorder. Hum Brain Mapp 29: 1265–1275 [PMC free article] [PubMed] [Google Scholar]
  • Calon F, Dridi M, Hornykiewicz O, Bedard PJ, Rajput AH, Di Paolo T 2004. Increased adenosine A2A receptors in the brain of Parkinson’s disease patients with dyskinesias. Brain 127: 1075–1084 [PubMed] [Google Scholar]
  • Carbon M, Ghilardi MF, Feigin A, Fukuda M, Silvestri G, Mentis MJ, Ghez C, Moeller JR, Eidelberg D 2003. Learning networks in health and Parkinson’s disease: Reproducibility and treatment effects. Hum Brain Mapp 19: 197–211 [PMC free article] [PubMed] [Google Scholar]
  • Carbon M, Ma Y, Barnes A, Dhawan V, Chaly T, Ghilardi MF, Eidelberg D 2004. Caudate nucleus: Influence of dopaminergic input on sequence learning and brain activation in Parkinsonism. Neuroimage 21: 1497–1507 [PubMed] [Google Scholar]
  • Carlsson T, Carta M, Munoz A, Mattsson B, Winkler C, Kirik D, Bjorklund A 2009. Impact of grafted serotonin and dopamine neurons on development of L-DOPA-induced dyskinesias in parkinsonian rats is determined by the extent of dopamine neuron degeneration. Brain 132: 319–335 [PubMed] [Google Scholar]
  • Carta M, Carlsson T, Kirik D, Bjorklund A 2007. Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain 130: 1819–1833 [PubMed] [Google Scholar]
  • Cenci MA, Lindgren HS 2007. Advances in understanding L-DOPA-induced dyskinesia. Curr Opin Neurobiol 17: 665–671 [PubMed] [Google Scholar]
  • Cenci MA, Lundblad M 2006. Post- versus presynaptic plasticity in L-DOPA-induced dyskinesia. J Neurochem 99: 381–392 [PubMed] [Google Scholar]
  • Colloby SJ, Pakrasi S, Firbank MJ, Perry EK, Piggott MA, Owens J, Wyper DJ, McKeith IG, Burn DJ, Williams ED, et al. 2006. In vivo SPECT imaging of muscarinic acetylcholine receptors using (R,R) 123I-QNB in dementia with Lewy bodies and Parkinson’s disease dementia. Neuroimage 33: 423–429 [PubMed] [Google Scholar]
  • Constantinescu R, Richard I, Kurlan R 2007. Levodopa responsiveness in disorders with parkinsonism: A review of the literature. Mov Disord 29: 29 [PubMed] [Google Scholar]
  • Cools R 2006. Dopaminergic modulation of cognitive function–implications for L-DOPA treatment in Parkinson's disease. Neurosci Biobehav Rev 30: 1–23 [PubMed] [Google Scholar]
  • Cools R, Stefanova E, Barker RA, Robbins TW, Owen AM 2002. Dopaminergic modulation of high-level cognition in Parkinson's disease: The role of the prefrontal cortex revealed by PET. Brain 125: 584–594 [PubMed] [Google Scholar]
  • Damoiseaux JS, Rombouts SA, Barkhof F, Scheltens P, Stam CJ, Smith SM, Beckmann CF 2006. Consistent resting-state networks across healthy subjects. Proc Natl Acad Sci 103: 13848–13853 [PMC free article] [PubMed] [Google Scholar]
  • de la Fuente-Fernandez R, Sossi V, Huang Z, Furtado S, Lu JQ, Calne DB, Ruth TJ, Stoessl AJ 2004. Levodopa-induced changes in synaptic dopamine levels increase with progression of Parkinson’s disease: Implications for dyskinesias. Brain 127: 2747–2754 [PubMed] [Google Scholar]
  • DeLong MR, Wichmann T 2007. Circuits and circuit disorders of the basal ganglia. Arch Neurol 64: 20–24 [PubMed] [Google Scholar]
  • Dentresangle C, Veyre L, Le Bars D, Pierre C, Lavenne F, Pollak P, Guerin J, Froment JC, Brousolle E 1999. Striatal D2 dopamine receptor status in Parkinson’s disease: An [18F]dopa and [11C]raclopride PET study. Mov Disord 14: 1025–1030 [PubMed] [Google Scholar]
  • Dhawan V, Eidelberg D 2006. PET imaging in Parkinson’s disease and other neurodegenerative disorders. In Neurobiology of disease (ed. Gilman S.), pp. 821–828 Academic Press, San Diego [Google Scholar]
  • Eckert T, Tang C, Eidelberg D 2007a. Assessment of the progression of Parkinson’s disease: A metabolic network approach. Lancet Neurol 6: 926–932 [PMC free article] [PubMed] [Google Scholar]
  • Eckert T, Van Laere K, Tang C, Lewis DE, Edwards C, Santens P, Eidelberg D 2007b. Quantification of Parkinson’s disease-related network expression with ECD SPECT. Eur J Nucl Med Mol Imaging 34: 496–501 [PubMed] [Google Scholar]
  • Eckert T, Tang C, Ma Y, Brown N, Lin T, Frucht S, Feigin A, Eidelberg D 2008. Abnormal metabolic networks in atypical parkinsonism. Mov Disord 23: 727–733 [PubMed] [Google Scholar]
  • Edison P, Rowe CC, Rinne JO, Ng S, Ahmed I, Kemppainen N, Villemagne VL, O’Keefe G, Nagren K, Chaudhury KR, et al. 2008. Amyloid load in Parkinson’s disease dementia and Lewy body dementia measured with [11C]PIB positron emission tomography. J Neurol Neurosurg Psychiatry 79: 1331–1338 [PubMed] [Google Scholar]
  • Edvinsson L, Krause D 2002. Catecholamines. In Cerebral blood flow and metabolism (ed. Edvinsson L, Krause D), pp. 191–211 Lippincott Williams & Wilkins, Philadelphia [Google Scholar]
  • Eidelberg D 2009. Metabolic brain networks in neurodegenerative disorders: A functional imaging approach. Trends Neurosci 32: 548–557 [PMC free article] [PubMed] [Google Scholar]
  • Eidelberg D, Moeller JR, Dhawan V, Sidtis JJ, Ginos JZ, Strother SC, Cedarbaum J, Greene P, Fahn S, Rottenberg DA 1990. The metabolic anatomy of Parkinson’s disease: Complementary [18F]fluorodeoxyglucose and [18F]fluorodopa positron emission tomographic studies. Mov Disord 5: 203–213 [PubMed] [Google Scholar]
  • Eidelberg D, Moeller JR, Dhawan V, Spetsieris P, Takikawa S, Ishikawa T, Chaly T, Robeson W, Margouleff D, Przedborski S, et al. 1994. The metabolic topography of parkinsonism. J Cereb Blood Flow Metab 14: 783–801 [PubMed] [Google Scholar]
  • Eidelberg D, Moeller JR, Ishikawa T, Dhawan V, Spetsieris P, Chaly T, Belakhlef A, Mandel F, Przedborski S, Fahn S 1995a. Early differential diagnosis of Parkinson’s disease with 18F-fluorodeoxyglucose and positron emission tomography. Neurology 45: 1995–2004 [PubMed] [Google Scholar]
  • Eidelberg D, Moeller JR, Ishikawa T, Dhawan V, Spetsieris P, Chaly T, Robeson W, Dahl JR, Margouleff D 1995b. Assessment of disease severity in parkinsonism with fluorine-18-fluorodeoxyglucose and PET. J Nucl Med 36: 378–383 [PubMed] [Google Scholar]
  • Eisensehr I, Linke R, Noachtar S, Schwarz J, Gildehaus FJ, Tatsch K 2000. Reduced striatal dopamine transporters in idiopathic rapid eye movement sleep behaviour disorder. Comparison with Parkinson’s disease and controls. Brain 123 (Pt 6): 1155–1160 [PubMed] [Google Scholar]
  • Emre M, Aarsland D, Albanese A, Byrne EJ, Deuschl G, De Deyn PP, Durif F, Kulisevsky J, van Laar T, Lees A, et al. 2004. Rivastigmine for dementia associated with Parkinson’s disease. New Engl J Med 351: 2509–2518 [PubMed] [Google Scholar]
  • Fabbrini G, Brotchie JM, Grandas F, Nomoto M, Goetz CG 2007. Levodopa-induced dyskinesias. Mov Disord 22: 1379–1389 [PubMed] [Google Scholar]
  • Feigin A, Fukuda M, Dhawan V, Przedborski S, Jackson-Lewis V, Mentis MJ, Moeller JR, Eidelberg D 2001. Metabolic correlates of levodopa response in Parkinson’s disease. Neurology 57: 2083–2088 [PubMed] [Google Scholar]
  • Feigin A, Antonini A, Fukuda M, De Notaris R, Benti R, Pezzoli G, Mentis MJ, Moeller JR, Eidelberg D 2002. Tc-99m ethylene cysteinate dimer SPECT in the differential diagnosis of parkinsonism. Mov Disord 17: 1265–1270 [PubMed] [Google Scholar]
  • Feigin A, Ghilardi MF, Carbon M, Edwards C, Fukuda M, Dhawan V, Margouleff C, Ghez C, Eidelberg D 2003. Effects of levodopa on motor sequence learning in Parkinson's disease. Neurology 60: 1744–1749 [PubMed] [Google Scholar]
  • Feigin A, Tang C, Ma Y, Mattis P, Zgaljardic D, Guttman M, Paulsen JS, Dhawan V, Eidelberg D 2007. Thalamic metabolism and symptom onset in preclinical Huntington’s disease. Brain 130: 2858–2867 [PMC free article] [PubMed] [Google Scholar]
  • Filippi L, Manni C, Pierantozzi M, Brusa L, Danieli R, Stanzione P, Schillaci O 2005. 123I-FP-CIT semi-quantitative SPECT detects preclinical bilateral dopaminergic deficit in early Parkinson’s disease with unilateral symptoms. Nucl Med Commun 26: 421–426 [PubMed] [Google Scholar]
  • Fodero-Tavoletti MT, Smith DP, McLean CA, Adlard PA, Barnham KJ, Foster LE, Leone L, Perez K, Cortes M, Culvenor JG, et al. 2007. In vitro characterization of Pittsburgh compound-B binding to Lewy bodies. J Neurosci 27: 10365–10371 [PMC free article] [PubMed] [Google Scholar]
  • Fodero-Tavoletti MT, Mulligan RS, Okamura N, Furumoto S, Rowe CC, Kudo Y, Masters CL, Cappai R, Yanai K, Villemagne VL 2009. In vitro characterisation of BF227 binding to α-synuclein/Lewy bodies. Eur J Pharmacol 617: 54–58 [PubMed] [Google Scholar]
  • Fox MD, Raichle ME 2007. Spontaneous fluctuations in brain activity observed with functional magnetic resonance imaging. Nat Rev Neurosci 8: 700–711 [PubMed] [Google Scholar]
  • Fox SH, Henry B, Hill MP, Peggs D, Crossman AR, Brotchie JM 2001. Neural mechanisms underlying peak-dose dyskinesia induced by levodopa and apomorphine are distinct: Evidence from the effects of the α(2) adrenoceptor antagonist idazoxan. Mov Disord 16: 642–650 [PubMed] [Google Scholar]
  • Fox SH, Lang AE, Brotchie JM 2006. Translation of nondopaminergic treatments for levodopa-induced dyskinesia from MPTP-lesioned nonhuman primates to phase IIa clinical studies: Keys to success and roads to failure. Mov Disord 21: 1578–1594 [PubMed] [Google Scholar]
  • Fujita M, Ichise M, Zoghbi SS, Liow JS, Ghose S, Vines DC, Sangare J, Lu JQ, Cropley VL, Iida H, et al. 2006. Widespread decrease of nicotinic acetylcholine receptors in Parkinson’s disease. Ann Neurol 59: 174–177 [PMC free article] [PubMed] [Google Scholar]
  • Fukuda M, Mentis M, Ghilardi MF, Dhawan V, Antonini A, Hammerstad J, Lozano AM, Lang A, Lyons K, Koller W, et al. 2001a. Functional correlates of pallidal stimulation for Parkinson’s disease. Ann Neurol 49: 155–164 [PubMed] [Google Scholar]
  • Fukuda M, Mentis MJ, Ma Y, Dhawan V, Antonini A, Lang AE, Lozano AM, Hammerstad J, Lyons K, Koller WC, et al. 2001b. Networks mediating the clinical effects of pallidal brain stimulation for Parkinson’s disease: A PET study of resting-state glucose metabolism. Brain 124: 1601–1609 [PubMed] [Google Scholar]
  • Fukuda M, Ghilardi MF, Carbon M, Dhawan V, Ma Y, Feigin A, Mentis MJ, Ghez C, Eidelberg D 2002. Pallidal stimulation for parkinsonism: Improved brain activation during sequence learning. Ann Neurol 52: 144–152 [PubMed] [Google Scholar]
  • Gai WP, Halliday GM, Blumbergs PC, Geffen LB, Blessing WW 1991. Substance P-containing neurons in the mesopontine tegmentum are severely affected in Parkinson’s disease. Brain 114 (Pt 5): 2253–2267 [PubMed] [Google Scholar]
  • Galvin JE, Lee VM, Trojanowski JQ 2001. Synucleinopathies: Clinical and pathological implications. Arch Neurol 58: 186–190 [PubMed] [Google Scholar]
  • Giladi N, Shabtai H, Gurevich T, Benbunan B, Anca M, Korczyn AD 2003. Rivastigmine (Exelon) for dementia in patients with Parkinson’s disease. Acta Neurol Scand 108: 368–373 [PubMed] [Google Scholar]
  • Gomperts SN, Rentz DM, Moran E, Becker JA, Locascio JJ, Klunk WE, Mathis CA, Elmaleh DR, Shoup T, Fischman AJ, et al. 2008. Imaging amyloid deposition in Lewy body diseases. Neurology 71: 903–910 [PMC free article] [PubMed] [Google Scholar]
  • Grady CL, Springer MV, Hongwanishkul D, McIntosh AR, Winocur G 2006. Age-related changes in brain activity across the adult lifespan. J Cogn Neurosci 18: 227–241 [PubMed] [Google Scholar]
  • Green J, McDonald WM, Vitek JL, Evatt M, Freeman A, Haber M, Bakay RA, Triche S, Sirockman B, DeLong MR 2002. Cognitive impairments in advanced PD without dementia. Neurology 59: 1320–1324 [PubMed] [Google Scholar]
  • Habeck C, Stern Y 2010. Multivariate data analysis for neuroimaging data: Overview and application to Alzheimer’s disease. Cell Biochem Biophys 58: 53–67 [PMC free article] [PubMed] [Google Scholar]
  • Habeck C, Krakauer JW, Ghez C, Sackeim HA, Eidelberg D, Stern Y, Moeller JR 2005. A new approach to spatial covariance modeling of functional brain imaging data: Ordinal trend analysis. Neural Comput 17: 1602–1645 [PubMed] [Google Scholar]
  • Habeck C, Foster NL, Perneczky R, Kurz A, Alexopoulos P, Koeppe RA, Drzezga A, Stern Y 2008. Multivariate and univariate neuroimaging biomarkers of Alzheimer’s disease. Neuroimage 40: 1503–1515 [PMC free article] [PubMed] [Google Scholar]
  • Hallett PJ, Brotchie JM 2007. Striatal delta opioid receptor binding in experimental models of Parkinson’s disease and dyskinesia. Mov Disord 22: 28–40 [PubMed] [Google Scholar]
  • Helmich RC, Derikx LC, Bakker M, Scheeringa R, Bloem BR, Toni I 2010. Spatial remapping of cortico-striatal connectivity in Parkinson’s disease. Cereb Cortex 20: 1175–1186 [PubMed] [Google Scholar]
  • Henriksen L, Boas J 1985. Regional cerebral blood flow in hemiparkinsonian patients. Emission computerized tomography of inhaled 133Xenon before and after levodopa. Acta Neurol Scand 71: 257–266 [PubMed] [Google Scholar]
  • Herholz K, Bauer B, Wienhard K, Kracht L, Mielke R, Lenz MO, Strotmann T, Heiss WD 2000. In-vivo measurements of regional acetylcholine esterase activity in degenerative dementia: Comparison with blood flow and glucose metabolism. J Neural Transm 107: 1457–1468 [PubMed] [Google Scholar]
  • Hershey T, Black KJ, Stambuk MK, Carl JL, McGee-Minnich LA, Perlmutter JS 1998. Altered thalamic response to levodopa in Parkinson’s patients with dopa-induced dyskinesias. Proc Natl Acad Sci 95: 12016–12021 [PMC free article] [PubMed] [Google Scholar]
  • Hershey T, Black KJ, Carl JL, McGee-Minnich L, Snyder AZ, Perlmutter JS 2003. Long term treatment and disease severity change brain responses to levodopa in Parkinson’s disease. J Neurol Neurosurg Psychiatry 74: 844–851 [PMC free article] [PubMed] [Google Scholar]
  • Hilker R, Thomas AV, Klein JC, Weisenbach S, Kalbe E, Burghaus L, Jacobs AH, Herholz K, Heiss WD 2005. Dementia in Parkinson disease: Functional imaging of cholinergic and dopaminergic pathways. Neurology 65: 1716–1722 [PubMed] [Google Scholar]
  • Hirano S, Asanuma K, Ma Y, Tang C, Feigin A, Dhawan V, Carbon M, Eidelberg D 2008. Dissociation of metabolic and neurovascular responses to levodopa in the treatment of Parkinson’s disease. J Neurosci 28: 4201–4209 [PMC free article] [PubMed] [Google Scholar]
  • Hirano S, Dhawan V, Eidelberg D 2010. PET Imaging in movement disorders. In Encyclopedia of movement disorders (ed. Kompoliti K, Verhagen L), pp. 452–461 Academic Press, New York [Google Scholar]
  • Hosokai Y, Nishio Y, Hirayama K, Takeda A, Ishioka T, Sawada Y, Suzuki K, Itoyama Y, Takahashi S, Fukuda H, et al. 2009. Distinct patterns of regional cerebral glucose metabolism in Parkinson’s disease with and without mild cognitive impairment. Mov Disord 24: 854–862 [PubMed] [Google Scholar]
  • Huang C, Mattis P, Tang C, Perrine K, Carbon M, Eidelberg D 2007a. Metabolic brain networks associated with cognitive function in Parkinson’s disease. Neuroimage 34: 714–723 [PMC free article] [PubMed] [Google Scholar]
  • Huang C, Tang C, Feigin A, Lesser M, Ma Y, Pourfar M, Dhawan V, Eidelberg D 2007b. Changes in network activity with the progression of Parkinson’s disease. Brain 130: 1834–1846 [PMC free article] [PubMed] [Google Scholar]
  • Huang C, Mattis P, Perrine K, Brown N, Dhawan V, Eidelberg D 2008. Metabolic abnormalities associated with mild cognitive impairment in Parkinson disease. Neurology 70: 1470–1477 [PMC free article] [PubMed] [Google Scholar]
  • Iadecola C 1998. Neurogenic control of the cerebral microcirculation: Is dopamine minding the store? Nat Neurosci 1: 263–265 [PubMed] [Google Scholar]
  • Irie T, Fukushi K, Namba H, Iyo M, Tamagami H, Nagatsuka S, Ikota N 1996. Brain acetylcholinesterase activity: Validation of a PET tracer in a rat model of Alzheimer’s disease. J Nucl Med 37: 649–655 [PubMed] [Google Scholar]
  • Isaias IU, Benti R, Cilia R, Canesi M, Marotta G, Gerundini P, Pezzoli G, Antonini A 2007. [123I]FP-CIT striatal binding in early Parkinson’s disease patients with tremor vs. akinetic-rigid onset. Neuroreport 18: 1499–1502 [PubMed] [Google Scholar]
  • Isaias IU, Marotta G, Hirano S, Canesi M, Benti R, Righini A, Tang C, Cilia R, Pezzoli G, Eidelberg D, et al. 2010. Imaging essential tremor. Mov Disord 25: 679–686 [PubMed] [Google Scholar]
  • Ito K, Nagano-Saito A, Kato T, Arahata Y, Nakamura A, Kawasumi Y, Hatano K, Abe Y, Yamada T, Kachi T, et al. 2002. Striatal and extrastriatal dysfunction in Parkinson’s disease with dementia: A 6-[18F]fluoro-L-dopa PET study. Brain 125: 1358–1365 [PubMed] [Google Scholar]
  • Jellinger K 1988. The pedunculopontine nucleus in Parkinson’s disease, progressive supranuclear palsy and Alzheimer’s disease. J Neurol Neurosurg Psychiatry 51: 540–543 [PMC free article] [PubMed] [Google Scholar]
  • Jellinger KA, Seppi K, Wenning GK, Poewe W 2002. Impact of coexistent Alzheimer pathology on the natural history of Parkinson’s disease. J Neural Transm 109: 329–339 [PubMed] [Google Scholar]
  • Johansson A, Savitcheva I, Forsberg A, Engler H, Langstrom B, Nordberg A, Askmark H 2008. [(11)C]-PIB imaging in patients with Parkinson’s disease: Preliminary results. Parkinsonism Relat Disord 14: 345–347 [PubMed] [Google Scholar]
  • Jokinen P, Bruck A, Aalto S, Forsback S, Parkkola R, Rinne JO 2009. Impaired cognitive performance in Parkinson’s disease is related to caudate dopaminergic hypofunction and hippocampal atrophy. Parkinsonism Relat Disord 15: 88–93 [PubMed] [Google Scholar]
  • Kas A, Bottlaender M, Gallezot JD, Vidailhet M, Villafane G, Gregoire MC, Coulon C, Valette H, Dolle F, Ribeiro MJ, et al. 2009. Decrease of nicotinic receptors in the nigrostriatal system in Parkinson’s disease. J Cereb Blood Flow Metab 29: 1601–1608 [PubMed] [Google Scholar]
  • Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergstrom M, Savitcheva I, Huang GF, Estrada S, et al. 2004. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann Neurol 55: 306–319 [PubMed] [Google Scholar]
  • Kobari M, Fukuuchi Y, Shinohara T, Obara K, Nogawa S 1995. Levodopa-induced local cerebral blood flow changes in Parkinson’s disease and related disorders. J Neurol Sci 128: 212–218 [PubMed] [Google Scholar]
  • Kuhl DE, Minoshima S, Fessler JA, Frey KA, Foster NL, Ficaro EP, Wieland DM, Koeppe RA 1996. In vivo mapping of cholinergic terminals in normal aging, Alzheimer’s disease, and Parkinson’s disease. Ann Neurol 40: 399–410 [PubMed] [Google Scholar]
  • Kwak Y, Muller ML, Bohnen NI, Dayalu P, Seidler RD 2010. Effect of dopaminergic medications on the time course of explicit motor sequence learning in Parkinson’s disease. J Neurophysiol 103: 942–949 [PubMed] [Google Scholar]
  • Lang AE 2011. A critical appraisal of the premotor symptoms of Parkinson’s disease: Potential usefulness in early diagnosis and design of neuroprotective trials. Mov Disord 26: 775–783 [PubMed] [Google Scholar]
  • Lim SY, Lang AE 2010. The nonmotor symptoms of Parkinson’s disease—An overview. Mov Disord 25 Suppl 1: S123–S130 [PubMed] [Google Scholar]
  • Lin TP, Carbon M, Tang C, Mogilner AY, Sterio D, Beric A, Dhawan V, Eidelberg D 2008. Metabolic correlates of subthalamic nucleus activity in Parkinson’s disease. Brain 131: 1373–1380 [PubMed] [Google Scholar]
  • Linazasoro G, Antonini A, Maguire RP, Leenders KL 2004. Pharmacological and PET studies in patients with Parkinson’s disease and a short duration-motor response: Implications in the pathophysiology of motor complications. J Neural Transm 111: 497–509 [PubMed] [Google Scholar]
  • Linazasoro G, Van Blercom N, Bergaretxe A, Inaki FM, Laborda E, Ruiz Ortega JA 2009. Levodopa-induced dyskinesias in Parkinson disease are independent of the extent of striatal dopaminergic denervation: A pharmacological and SPECT study. Clin Neuropharmacol 32: 326–329 [PubMed] [Google Scholar]
  • Lindgren HS, Andersson DR, Lagerkvist S, Nissbrandt H, Cenci MA 2010. L-DOPA-induced dopamine efflux in the striatum and the substantia nigra in a rat model of Parkinson’s disease: Temporal and quantitative relationship to the expression of dyskinesia. J Neurochem 112: 1465–1476 [PubMed] [Google Scholar]
  • Lozza C, Baron JC, Eidelberg D, Mentis MJ, Carbon M, Marie RM 2004. Executive processes in Parkinson’s disease: FDG-PET and network analysis. Hum Brain Mapp 22: 236–245 [PMC free article] [PubMed] [Google Scholar]
  • Lustig C, Snyder AZ, Bhakta M, O’Brien KC, McAvoy M, Raichle ME, Morris JC, Buckner RL 2003. Functional deactivations: Change with age and dementia of the Alzheimer type. Proc Natl Acad Sci 100: 14504–14509 [PMC free article] [PubMed] [Google Scholar]
  • Ma Y, Eidelberg D 2007. Functional imaging of cerebral blood flow and glucose metabolism in Parkinson’s disease and Huntington’s disease. Mol Imaging Biol 9: 223–233 [PMC free article] [PubMed] [Google Scholar]
  • Ma Y, Tang C, Spetsieris P, Dhawan V, Eidelberg D 2007. Abnormal metabolic network activity in Parkinson’s disease: Test-retest reproducibility. J Cereb Blood Flow Metab 27: 597–605 [PMC free article] [PubMed] [Google Scholar]
  • Ma Y, Huang C, Dyke JP, Pan H, Alsop D, Feigin A, Eidelberg D 2010. Parkinson’s disease spatial covariance pattern: Noninvasive quantification with perfusion MRI. J Cereb Blood Flow Metab 30: 505–509 [PMC free article] [PubMed] [Google Scholar]
  • Ma Y, Peng S, Spetsieris P, Sossi V, Eidelberg D, Doudet DJ 2011. Abnormal metabolic brain networks in a non-human primate model of parkinsonism. J Cereb Blood Flow Metab 10.1038/jcbfm.2011.166 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Maetzler W, Reimold M, Liepelt I, Solbach C, Leyhe T, Schweitzer K, Eschweiler GW, Mittelbronn M, Gaenslen A, Uebele M, et al. 2008. [11C]PIB binding in Parkinson’s disease dementia. Neuroimage 39: 1027–1033 [PubMed] [Google Scholar]
  • Maetzler W, Liepelt I, Reimold M, Reischl G, Solbach C, Becker C, Schulte C, Leyhe T, Keller S, Melms A, et al. 2009. Cortical PIB binding in Lewy body disease is associated with Alzheimer-like characteristics. Neurobiol Dis 34: 107–112 [PubMed] [Google Scholar]
  • Margulies DS, Bottger J, Long X, Lv Y, Kelly C, Schafer A, Goldhahn D, Abbushi A, Milham MP, Lohmann G, et al. 2010. Resting developments: A review of fMRI post-processing methodologies for spontaneous brain activity. MAGMA 23: 289–307 [PubMed] [Google Scholar]
  • Marie RM, Barre L, Dupuy B, Viader F, Defer G, Baron JC 1999. Relationships between striatal dopamine denervation and frontal executive tests in Parkinson’s disease. Neurosci Lett 260: 77–80 [PubMed] [Google Scholar]
  • Mattay VS, Tessitore A, Callicott JH, Bertolino A, Goldberg TE, Chase TN, Hyde TM, Weinberger DR 2002. Dopaminergic modulation of cortical function in patients with Parkinson's disease. Ann Neurol 51: 156–164 [PubMed] [Google Scholar]
  • Mattis PJ, Tang CC, Ma Y, Dhawan V, Eidelberg D 2011. Network correlates of the cognitive response to levodopa in Parkinson disease. Neurology 77: 858–865 [PMC free article] [PubMed] [Google Scholar]
  • Melzer TR, Watts R, MacAskill MR, Pearson JF, Rueger S, Pitcher TL, Livingston L, Graham C, Keenan R, Shankaranarayanan A, et al. 2011. Arterial spin labelling reveals an abnormal cerebral perfusion pattern in Parkinson’s disease. Brain 134: 845–855 [PMC free article] [PubMed] [Google Scholar]
  • Mentis MJ, McIntosh AR, Perrine K, Dhawan V, Berlin B, Feigin A, Edwards C, Mattis P, Eidelberg D 2002. Relationships among the metabolic patterns that correlate with mnemonic, visuospatial, and mood symptoms in Parkinson’s disease. Am J Psychiatry 159: 746–754 [PubMed] [Google Scholar]
  • Meyer PM, Strecker K, Kendziorra K, Becker G, Hesse S, Woelpl D, Hensel A, Patt M, Sorger D, Wegner F, et al. 2009. Reduced α4β2*-nicotinic acetylcholine receptor binding and its relationship to mild cognitive and depressive symptoms in Parkinson disease. Arch Gen Psychiatry 66: 866–877 [PubMed] [Google Scholar]
  • Moeller JR, Nakamura T, Mentis MJ, Dhawan V, Spetsieres P, Antonini A, Missimer J, Leenders KL, Eidelberg D 1999. Reproducibility of regional metabolic covariance patterns: Comparison of four populations. J Nucl Med 40: 1264–1269 [PubMed] [Google Scholar]
  • Montastruc JL, Celsis P, Agniel A, Demonet JF, Doyon B, Puel M, Marc-Vergnes JP, Rascol A 1987. Levodopa-induced regional cerebral blood flow changes in normal volunteers and patients with Parkinson’s disease. Lack of correlation with clinical or neuropsychological improvements. Mov Disord 2: 279–289 [PubMed] [Google Scholar]
  • Morrish PK, Sawle GV, Brooks DJ 1996. Regional changes in [18F]dopa metabolism in the striatum in Parkinson’s disease. Brain 119 (Pt 6): 2097–2103 [PubMed] [Google Scholar]
  • Muller U, Wachter T, Barthel H, Reuter M, von Cramon DY 2000. Striatal [123I]β-CIT SPECT and prefrontal cognitive functions in Parkinson’s disease. J Neural Transm 107: 303–319 [PubMed] [Google Scholar]
  • Mure H, Hirano S, Tang CC, Isaias IU, Antonini A, Ma Y, Dhawan V, Eidelberg D 2011. Parkinson’s disease tremor-related metabolic network: Characterization, progression, and treatment effects. Neuroimage 54: 1244–1253 [PMC free article] [PubMed] [Google Scholar]
  • Nagano-Saito A, Liu J, Doyon J, Dagher A 2009. Dopamine modulates default mode network deactivation in elderly individuals during the Tower of London task. Neurosci Lett 458: 1–5 [PubMed] [Google Scholar]
  • Niethammer M, Eidelberg D 2012. Neuroimaging studies in the evaluation of patients with movement disorders. In Tremor, Parkinson’s disease and related movement disorders (ed. LeWitt PA.). Cambridge University Press, Cambridge [Google Scholar]
  • Niethammer M, Feigin A 2011. Imaging applications to Parkinson’s disease clinical trials. In Imaging in Parkinson’s disease (ed. Eidelberg D.), pp. 181–190 Oxford University Press, New York [Google Scholar]
  • Nurmi E, Ruottinen HM, Bergman J, Haaparanta M, Solin O, Sonninen P, Rinne JO 2001. Rate of progression in Parkinson’s disease: A 6-[18F]fluoro-L-dopa PET study. Mov Disord 16: 608–615 [PubMed] [Google Scholar]
  • O’Brien JT, Colloby S, Fenwick J, Williams ED, Firbank M, Burn D, Aarsland D, McKeith IG 2004. Dopamine transporter loss visualized with FP-CIT SPECT in the differential diagnosis of dementia with Lewy bodies. Arch Neurol 61: 919–925 [PubMed] [Google Scholar]
  • O’Brien JT, Colloby SJ, Pakrasi S, Perry EK, Pimlott SL, Wyper DJ, McKeith IG, Williams ED 2008. Nicotinic α4β2 receptor binding in dementia with Lewy bodies using 123I-5IA-85380 SPECT demonstrates a link between occipital changes and visual hallucinations. Neuroimage 40: 1056–1063 [PubMed] [Google Scholar]
  • Oishi N, Hashikawa K, Yoshida H, Ishizu K, Ueda M, Kawashima H, Saji H, Fukuyama H 2007. Quantification of nicotinic acetylcholine receptors in Parkinson’s disease with (123)I-5IA SPECT. J Neurol Sci 256: 52–60 [PubMed] [Google Scholar]
  • Pavese N, Evans AH, Tai YF, Hotton G, Brooks DJ, Lees AJ, Piccini P 2006. Clinical correlates of levodopa-induced dopamine release in Parkinson disease: A PET study. Neurology 67: 1612–1617 [PubMed] [Google Scholar]
  • Pavese N, Rivero-Bosch M, Lewis SJ, Whone AL, Brooks DJ 2011. Progression of monoaminergic dysfunction in Parkinson’s disease: A longitudinal 18F-dopa PET study. Neuroimage 56: 1463–1468 [PubMed] [Google Scholar]
  • Peng S, Ma Y, Flores J, Doudet DJ, Eidelberg D 2010. Abnormal topography of cerebral glucose metabolism in parkinsonian macaques. J Nucl Med 51: 1753 [Google Scholar]
  • Peppard RF, Martin WR, Clark CM, Carr GD, McGeer PL, Calne DB 1990. Cortical glucose metabolism in Parkinson’s and Alzheimer’s disease. J Neurosci Res 27: 561–568 [PubMed] [Google Scholar]
  • Piccini P, Weeks RA, Brooks DJ 1997. Alterations in opioid receptor binding in Parkinson’s disease patients with levodopa-induced dyskinesias. Ann Neurol 42: 720–726 [PubMed] [Google Scholar]
  • Piccini P, Burn DJ, Ceravolo R, Maraganore D, Brooks DJ 1999. The role of inheritance in sporadic Parkinson’s disease: Evidence from a longitudinal study of dopaminergic function in twins. Ann Neurol 45: 577–582 [PubMed] [Google Scholar]
  • Pimlott SL, Piggott M, Owens J, Greally E, Court JA, Jaros E, Perry RH, Perry EK, Wyper D 2004. Nicotinic acetylcholine receptor distribution in Alzheimer’s disease, dementia with Lewy bodies, Parkinson’s disease, and vascular dementia: In vitro binding study using 5-[(125)i]-a-85380. Neuropsychopharmacology 29: 108–116 [PubMed] [Google Scholar]
  • Pirker W 2003. Correlation of dopamine transporter imaging with parkinsonian motor handicap: How close is it? Mov Disord 18 Suppl 7: S43–S51 [PubMed] [Google Scholar]
  • Ponsen MM, Stoffers D, Booij J, van Eck-Smit BL, Wolters E, Berendse HW 2004. Idiopathic hyposmia as a preclinical sign of Parkinson’s disease. Ann Neurol 56: 173–181 [PubMed] [Google Scholar]
  • Poston KL 2011. Clinical applications. In Imaging in Parkinson’s disease (ed. Eidelberg D.), pp. 191–202 Oxford University Press, New York [Google Scholar]
  • Pourfar M, Tang C, Lin T, Dhawan V, Kaplitt MG, Eidelberg D 2009. Assessing the microlesion effect of subthalamic deep brain stimulation surgery with FDG PET. J Neurosurg 110: 1278–1282 [PubMed] [Google Scholar]
  • Priori A, Foffani G, Pesenti A, Tamma F, Bianchi AM, Pellegrini M, Locatelli M, Moxon KA, Villani RM 2004. Rhythm-specific pharmacological modulation of subthalamic activity in Parkinson’s disease. Exp Neurol 189: 369–379 [PubMed] [Google Scholar]
  • Raichle ME, Snyder AZ 2007. A default mode of brain function: A brief history of an evolving idea. Neuroimage 37: 1083–1090; discussion 1097–1089 [PubMed] [Google Scholar]
  • Raichle ME, MacLeod AM, Snyder AZ, Powers WJ, Gusnard DA, Shulman GL 2001. A default mode of brain function. Proc Natl Acad Sci 98: 676–682 [PMC free article] [PubMed] [Google Scholar]
  • Rajput AH, Fenton ME, Birdi S, Macaulay R, George D, Rozdilsky B, Ang LC, Senthilselvan A, Hornykiewicz O 2002. Clinical-pathological study of levodopa complications. Mov Disord 17: 289–296 [PubMed] [Google Scholar]
  • Rakshi JS, Uema T, Ito K, Bailey DL, Morrish PK, Ashburner J, Dagher A, Jenkins IH, Friston KJ, Brooks DJ 1999. Frontal, midbrain and striatal dopaminergic function in early and advanced Parkinson’s disease A 3D [(18)F]dopa-PET study. Brain 122 (Pt 9): 1637–1650 [PubMed] [Google Scholar]
  • Rascol O, Sabatini U, Brefel C, Fabre N, Rai S, Senard JM, Celsis P, Viallard G, Montastruc JL, Chollet F 1998. Cortical motor overactivation in parkinsonian patients with L-dopa-induced peak-dose dyskinesia. Brain 121 (Pt 3): 527–533 [PubMed] [Google Scholar]
  • Rascol O, Arnulf I, Peyro-Saint Paul H, Brefel-Courbon C, Vidailhet M, Thalamas C, Bonnet AM, Descombes S, Bejjani B, Fabre N, et al. 2001. Idazoxan, an α-2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson’s disease. Mov Disord 16: 708–713 [PubMed] [Google Scholar]
  • Ribeiro MJ, Vidailhet M, Loc’h C, Dupel C, Nguyen JP, Ponchant M, Dolle F, Peschanski M, Hantraye P, Cesaro P, et al. 2002. Dopaminergic function and dopamine transporter binding assessed with positron emission tomography in Parkinson disease. Arch Neurol 59: 580–586 [PubMed] [Google Scholar]
  • Rinne JO, Portin R, Ruottinen H, Nurmi E, Bergman J, Haaparanta M, Solin O 2000. Cognitive impairment and the brain dopaminergic system in Parkinson disease: [18F]fluorodopa positron emission tomographic study. Arch Neurol 57: 470–475 [PubMed] [Google Scholar]
  • Rioux L, Frohna PA, Joyce JN, Schneider JS 1997. The effects of chronic levodopa treatment on pre- and postsynaptic markers of dopaminergic function in striatum of parkinsonian monkeys. Mov Disord 12: 148–158 [PubMed] [Google Scholar]
  • Ruberg M, Rieger F, Villageois A, Bonnet AM, Agid Y 1986. Acetylcholinesterase and butyrylcholinesterase in frontal cortex and cerebrospinal fluid of demented and non-demented patients with Parkinson’s disease. Brain Res 362: 83–91 [PubMed] [Google Scholar]
  • Schmaljohann J, Gundisch D, Minnerop M, Bucerius J, Joe A, Reinhardt M, Guhlke S, Biersack HJ, Wullner U 2006. In vitro evaluation of nicotinic acetylcholine receptors with 2-[18F]F-A85380 in Parkinson’s disease. Nucl Med Biol 33: 305–309 [PubMed] [Google Scholar]
  • Shimada H, Hirano S, Shinotoh H, Aotsuka A, Sato K, Tanaka N, Ota T, Asahina M, Fukushi K, Kuwabara S, et al. 2009. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET. Neurology 73: 273–278 [PubMed] [Google Scholar]
  • Shinotoh H, Namba H, Yamaguchi M, Fukushi K, Nagatsuka S, Iyo M, Asahina M, Hattori T, Tanada S, Irie T 1999. Positron emission tomographic measurement of acetylcholinesterase activity reveals differential loss of ascending cholinergic systems in Parkinson’s disease and progressive supranuclear palsy. Ann Neurol 46: 62–69 [PubMed] [Google Scholar]
  • Smid LM, Vovko TD, Popovic M, Petric A, Kepe V, Barrio JR, Vidmar G, Bresjanac M 2006. The 2,6-disubstituted naphthalene derivative FDDNP labeling reliably predicts Congo red birefringence of protein deposits in brain sections of selected human neurodegenerative diseases. Brain Pathol 16: 124–130 [PMC free article] [PubMed] [Google Scholar]
  • Smith LA, Jackson MJ, Hansard MJ, Maratos E, Jenner P 2003. Effect of pulsatile administration of levodopa on dyskinesia induction in drug-naive MPTP-treated common marmosets: Effect of dose, frequency of administration, and brain exposure. Mov Disord 18: 487–495 [PubMed] [Google Scholar]
  • Snow BJ, Tooyama I, McGeer EG, Yamada T, Calne DB, Takahashi H, Kimura H 1993. Human positron emission tomographic [18F]fluorodopa studies correlate with dopamine cell counts and levels. Ann Neurol 34: 324–330 [PubMed] [Google Scholar]
  • Sommer U, Hummel T, Cormann K, Mueller A, Frasnelli J, Kropp J, Reichmann H 2004. Detection of presymptomatic Parkinson’s disease: Combining smell tests, transcranial sonography, and SPECT. Mov Disord 19: 1196–1202 [PubMed] [Google Scholar]
  • Sossi V, de La Fuente-Fernandez R, Holden JE, Doudet DJ, McKenzie J, Stoessl AJ, Ruth TJ 2002. Increase in dopamine turnover occurs early in Parkinson’s disease: Evidence from a new modeling approach to PET 18 F-fluorodopa data. J Cereb Blood Flow Metab 22: 232–239 [PubMed] [Google Scholar]
  • Sossi V, de la Fuente-Fernandez R, Holden JE, Schulzer M, Ruth TJ, Stoessl J 2004. Changes of dopamine turnover in the progression of Parkinson’s disease as measured by positron emission tomography: Their relation to disease-compensatory mechanisms. J Cereb Blood Flow Metab 24: 869–876 [PubMed] [Google Scholar]
  • Sossi V, de la Fuente-Fernandez R, Schulzer M, Adams J, Stoessl J 2006. Age-related differences in levodopa dynamics in Parkinson’s: Implications for motor complications. Brain 129: 1050–1058 [PubMed] [Google Scholar]
  • Spetsieris PG, Eidelberg D 2011. Scaled subprofile modeling of resting state imaging data in Parkinson’s disease: Methodological issues. Neuroimage 54: 2899–2914 [PMC free article] [PubMed] [Google Scholar]
  • Spetsieris P, Ma Y, Dhawan V, Moeller JR, Eidelberg D 2006. Highly automated computer-aided diagnosis of neurological disorders using functional brain imaging. Proc SPIE: Medical Imaging 6144: 61445M [Google Scholar]
  • Spetsieris PG, Ma Y, Dhawan V, Eidelberg D 2009. Differential diagnosis of parkinsonian syndromes using PCA-based functional imaging features. Neuroimage 45: 1241–1252 [PubMed] [Google Scholar]
  • Stiasny-Kolster K, Doerr Y, Moller JC, Hoffken H, Behr TM, Oertel WH, Mayer G 2005. Combination of “idiopathic” REM sleep behaviour disorder and olfactory dysfunction as possible indicator for α-synucleinopathy demonstrated by dopamine transporter FP-CIT-SPECT. Brain 128: 126–137 [PubMed] [Google Scholar]
  • Tang CC, Poston KL, Dhawan V, Eidelberg D 2010a. Abnormalities in metabolic network activity precede the onset of motor symptoms in Parkinson’s disease. J Neurosci 30: 1049–1056 [PMC free article] [PubMed] [Google Scholar]
  • Tang CC, Poston KL, Eckert T, Feigin A, Frucht S, Gudesblatt M, Dhawan V, Lesser M, Vonsattel JP, Fahn S, et al. 2010b. Differential diagnosis of parkinsonism: A metabolic imaging study using pattern analysis. Lancet Neurol 9: 149–158 [PMC free article] [PubMed] [Google Scholar]
  • Tang CC, Niethammer M, Gerber NJ, Mattis P, Ma Y, Eidelberg D 2011. Expression of a cognitive metabolic network in Parkinson’s disease correlates with dopaminergic function in caudate nucleus. Neurology 76: A489–A490 [Google Scholar]
  • Thobois S, Jahanshahi M, Pinto S, Frackowiak R, Limousin-Dowsey P 2004. PET and SPECT functional imaging studies in Parkinsonian syndromes: From the lesion to its consequences. Neuroimage 23: 1–16 [PubMed] [Google Scholar]
  • Tinaz S, Schendan HE, Stern CE 2008. Fronto-striatal deficit in Parkinson’s disease during semantic event sequencing. Neurobiol Aging 29: 397–407 [PMC free article] [PubMed] [Google Scholar]
  • Tolosa E, Gaig C, Santamaria J, Compta Y 2009. Diagnosis and the premotor phase of Parkinson disease. Neurology 72: S12–S20 [PubMed] [Google Scholar]
  • Troiano AR, de la Fuente-Fernandez R, Sossi V, Schulzer M, Mak E, Ruth TJ, Stoessl AJ 2009. PET demonstrates reduced dopamine transporter expression in PD with dyskinesias. Neurology 72: 1211–1216 [PubMed] [Google Scholar]
  • Trost M, Su S, Su P, Yen RF, Tseng HM, Barnes A, Ma Y, Eidelberg D 2006. Network modulation by the subthalamic nucleus in the treatment of Parkinson’s disease. Neuroimage 31: 301–307 [PMC free article] [PubMed] [Google Scholar]
  • Tuor UI, Edvinsson L, McCulloch J 1986. Catecholamines and the relationship between cerebral blood flow and glucose use. Am J Physiol 251: H824–H833 [PubMed] [Google Scholar]
  • Turjanski N, Lees AJ, Brooks DJ 1997. In vivo studies on striatal dopamine D1 and D2 site binding in L-dopa-treated Parkinson’s disease patients with and without dyskinesias. Neurology 49: 717–723 [PubMed] [Google Scholar]
  • van Beilen M, Portman AT, Kiers HA, Maguire RP, Kaasinen V, Koning M, Pruim J, Leenders KL 2008. Striatal FDOPA uptake and cognition in advanced non-demented Parkinson’s disease: A clinical and FDOPA-PET study. Parkinsonism Relat Disord 14: 224–228 [PubMed] [Google Scholar]
  • van de Ven VG, Formisano E, Prvulovic D, Roeder CH, Linden DE 2004. Functional connectivity as revealed by spatial independent component analysis of fMRI measurements during rest. Hum Brain Mapp 22: 165–178 [PMC free article] [PubMed] [Google Scholar]
  • van Eimeren T, Monchi O, Ballanger B, Strafella AP 2009. Dysfunction of the default mode network in Parkinson disease: A functional magnetic resonance imaging study. Arch Neurol 66: 877–883 [PMC free article] [PubMed] [Google Scholar]
  • Vingerhoets FJ, Snow BJ, Lee CS, Schulzer M, Mak E, Calne DB 1994. Longitudinal fluorodopa positron emission tomographic studies of the evolution of idiopathic parkinsonism. Ann Neurol 36: 759–764 [PubMed] [Google Scholar]
  • Vingerhoets FJ, Schulzer M, Calne DB, Snow BJ 1997. Which clinical sign of Parkinson’s disease best reflects the nigrostriatal lesion? Ann Neurol 41: 58–64 [PubMed] [Google Scholar]
  • Westin JE, Lindgren HS, Gardi J, Nyengaard JR, Brundin P, Mohapel P, Cenci MA 2006. Endothelial proliferation and increased blood-brain barrier permeability in the basal ganglia in a rat model of 3,4-dihydroxyphenyl-L-alanine-induced dyskinesia. J Neurosci 26: 9448–9461 [PMC free article] [PubMed] [Google Scholar]
  • Whitehouse PJ, Hedreen JC, White CL III, Price DL 1983. Basal forebrain neurons in the dementia of Parkinson disease. Ann Neurol 13: 243–248 [PubMed] [Google Scholar]
  • Whitehouse PJ, Martino AM, Wagster MV, Price DL, Mayeux R, Atack JR, Kellar KJ 1988. Reductions in [3H]nicotinic acetylcholine binding in Alzheimer’s disease and Parkinson’s disease: An autoradiographic study. Neurology 38: 720–723 [PubMed] [Google Scholar]
  • Whone AL, Watts RL, Stoessl AJ, Davis M, Reske S, Nahmias C, Lang AE, Rascol O, Ribeiro MJ, Remy P, et al. 2003. Slower progression of Parkinson’s disease with ropinirole versus levodopa: The REAL-PET study. Ann Neurol 54: 93–101 [PubMed] [Google Scholar]
  • Wu T, Long X, Zang Y, Wang L, Hallett M, Li K, Chan P 2009. Regional homogeneity changes in patients with Parkinson’s disease. Hum Brain Mapp 30: 1502–1510 [PMC free article] [PubMed] [Google Scholar]
  • Ye L, Velasco A, Fraser G, Beach TG, Sue L, Osredkar T, Libri V, Spillantini MG, Goedert M, Lockhart A 2008. In vitro high affinity α-synuclein binding sites for the amyloid imaging agent PIB are not matched by binding to Lewy bodies in postmortem human brain. J Neurochem 105: 1428–1437 [PMC free article] [PubMed] [Google Scholar]
  • Yong SW, Yoon JK, An YS, Lee PH 2007. A comparison of cerebral glucose metabolism in Parkinson’s disease, Parkinson’s disease dementia and dementia with Lewy bodies. Eur J Neurol 14: 1357–1362 [PubMed] [Google Scholar]
  • Zang Y, Jiang T, Lu Y, He Y, Tian L 2004. Regional homogeneity approach to fMRI data analysis. Neuroimage 22: 394–400 [PubMed] [Google Scholar]