Skip to main content
Erschienen in: BMC Medicine 1/2023

Open Access 01.12.2023 | Research article

Current coffee consumption is associated with decreased striatal dopamine transporter availability in Parkinson’s disease patients and healthy controls

verfasst von: Chao Wang, Cheng Zhou, Tao Guo, Yeerfan Jiaerken, Siyu Yang, Xiaopei Xu, Ling Hu, Peiyu Huang, Xiaojun Xu, Minming Zhang

Erschienen in: BMC Medicine | Ausgabe 1/2023

Abstract

Background

Coffee is the most widely consumed psychostimulant worldwide. Emerging evidence indicates that coffee consumption habit significantly reduces the risk of developing Parkinson’s disease (PD). However, the effect of coffee consumption on nigrostriatal dopaminergic neurodegeneration is still largely unknown. We therefore aim to investigate the role of coffee consumption in nigrostriatal dopaminergic neurodegeneration using dopamine transporter (DAT) imaging in PD and healthy controls (HC).

Methods

A total of 138 PD patients and 75 HC with questionnaires about coffee consumption, and DAT scans were recruited from the Parkinson’s Progression Markers Initiative cohort. Demographic, clinical, and striatal DAT characteristics were compared across subgroups of current, former, and never coffee consumers in PD and HC, respectively. Furthermore, partial correlation analyses were performed to determine whether there was a relationship between coffee cups consumed per day and striatal DAT characteristics in each striatal region. In addition, the factors that may have influenced the loss of nigrostriatal dopaminergic neurons were included in multiple linear regression analyses to identify significant contributing factors to DAT availability in each striatal region.

Results

PD patients had lower DAT availability in each striatal region than HC (p < 0.001). In PD patients, there were significant differences in DAT availability in the caudate (p = 0.008, Bonferroni corrected) across three PD subgroups. Specifically, post hoc tests showed that current coffee consumers had significantly lower DAT availability in the caudate than former coffee consumers (p = 0.01) and never coffee consumers (p = 0.022). In HC, there were significant differences in DAT availability in the caudate (p = 0.031, Bonferroni uncorrected) across three HC subgroups. Specifically, post hoc tests showed that current coffee consumers had significantly lower DAT availability in the caudate than former coffee consumers (p = 0.022). Moreover, correlation analysis revealed that cups per day were negatively correlated with DAT availability in the caudate in current consumers of PD patients (r =  − 0.219, p = 0.047). In addition, multiple linear regression analyses showed that current coffee consumption remained an independent predictor of decreased DAT availability in the caudate in PD patients and HC.

Conclusions

This study demonstrates that current coffee consumption is associated with decreased striatal DAT availability in the caudate. However, the effects of caffeine on striatal DAT may fade and disappear after quitting coffee consumption.

Trial registration

ClinicalTrials.gov, NCT01141023.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-023-02994-5.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ANOVA
One-way analysis of variance
DAT
Dopamine transporter
ESS
Epworth Sleepiness Scale
GDS
Geriatric Depression Scale
HC
Healthy controls
HC-CC
Current consumers of healthy controls
HC-FC
Former consumers of healthy controls
HC-NC
Never consumers of healthy controls
H-Y
Hoehn and Yahr
MDS-UPDRS
Movement Disorders Society Unified Parkinson’s Disease Rating Scale
MNI
Montreal Neurologic Institute
MOCA
Montreal Cognitive Assessment
MPTP
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine
PD
Parkinson’s disease
PD-CC
Current consumers of Parkinson’s disease patients
PD-FC
Former consumers of Parkinson’s disease patients
PD-NC
Never consumers of Parkinson’s disease patients
PPMI
Parkinson’s Progression Markers Initiative
QUIP
Questionnaire for Impulsive-Compulsive Disorders in Parkinson’s Disease
ROIs
Regions of interest
SBR
Striatal binding ratios
SCOPA-AUT
Scale for Outcomes for Parkinson’s Disease–autonomic function
SPECT
Single-photon emission computed tomography
STAI
State and Trait Anxiety Scale
UPSIT
University of Pennsylvania Smell Identification Test

Background

Coffee is the most widely consumed psychostimulant worldwide [1]. After consumption, caffeine is rapidly absorbed through the gastrointestinal tract to the blood and then to the brain [2], leading to its short-term effects of causing alertness and reducing fatigue [3]. Long-term coffee consumption is associated with improved cardiovascular risk factors, improved asthma control, and lower liver disease risks and all-cause mortality [4]. Coffee consumption habit appears to attenuate the burden of some neurodegenerative diseases. Both retrospective and prospective epidemiological studies demonstrate that coffee consumption habit significantly reduces the risk of developing Parkinson’s disease (PD) [58], which is the second most common neurodegenerative disorder that affects 2–3% of elderly people > 65 years old worldwide [9]. Neuronal loss in the substantia nigra which causes a deficit of striatal dopamine and intracellular protein (α-synuclein) accumulation that leads to the nigrostriatal pathway impairment are the neuropathological hallmarks of PD [10]. The action of caffeine on the dopaminergic system is responsible for enhancing motor activity and exerting an antidyskinetic effect [11, 12]. There is evidence that coffee consumption is associated with reduced brain functional connectivity network related to somatosensory, motor, and emotional processing [13]. However, the effect of coffee consumption on nigrostriatal dopaminergic neurodegeneration is still largely unknown.
Dopamine transporter (DAT) is expressed in the dopaminergic neurons and clears the free dopamine released into the synaptic cleft [14]. In PD, the loss of dopaminergic neurons results in a substantial reduction of the DAT binding and dopamine levels [15]. Evidence indicates that caffeine acts as an adenosine receptor antagonist to interfere with dopaminergic transmission and enhance dopamine release with possible secondary reduced DAT availability [16]. In a mouse model of PD, exposure to caffeine significantly attenuates the loss of striatal DAT [17]. However, data are scarce from large, controlled human studies investigating the association between coffee consumption and striatal DAT availability in both PD patients and healthy controls (HC).
This study aimed to investigate whether coffee consumption was associated with the change in striatal DAT availability in PD patients and HC. Therefore, we systematically compared baseline striatal DAT availability across different coffee consumer subgroups (current, former, and never coffee consumers) in a large sample of newly diagnosed drug-naïve PD patients and HC enrolled from the Parkinson’s Progression Markers Initiative (PPMI) cohort.

Methods

Participants

Data used in this study were all obtained from the Parkinson’s Progression Markers Initiative (PPMI) database (http://​www.​ppmi-info.​org) on July 1, 2022. PPMI is an ongoing observational, international, multicenter cohort study in a large cohort (ClincalTrials.gov NCT01141023). The study of the PPMI database aimed to identify clinical, imaging, genetic, and biospecimen biomarkers of PD progression. In the PPMI study, de novo PD patients were prospectively enrolled and exhibited presynaptic dopaminergic terminal loss as confirmed by DAT imaging. Study protocols and manuals are available on the PPMI website. The data sourcing of coffee consumption questionnaires was obtained from PPMI FOUND. PPMI FOUND was designed to enhance retention and minimize loss to follow-up of persons enrolled in PPMI, by providing a parallel longitudinal assessment method using Internet, telephone, and/or mail contacts with PPMI participants. The coffee consumption data were retrospectively collected. Although there were > 400 de novo PD patients in PPMI, only a relatively small subset of the PD and HC who completed questionnaires on caffeine consumption were included in this study.
In all, 139 de novo PD patients and 75 HC with DAT scans and coffee consumption questionnaires were enrolled in this study. Coffee consumers were defined as people who have regularly drunk caffeinated coffee at least once per week for 6 months or longer and classified as “current” if they currently do and “former” if they no longer do. One PD patient who did not know/prefer not to answer the question “Do you currently drink caffeinated coffee?” was excluded. Therefore, this study finally included 138 PD patients and 75 HC. Specifically, PD patients consisted of 88 current consumers (PD-CC), 18 former consumers (PD-FC), and 32 never consumers (PD-NC), and HC consisted of 55 current consumers (HC-CC), 9 former consumers (HC-FC), and 11 never consumers (HC-NC). The participants’ flowchart is shown in Fig. 1.

Clinical evaluation

All participants in the PPMI cohort received the standard test battery of assessments. In addition to the demographic variables (age, gender, and education), the clinical variables were collected, including the Hoehn and Yahr (H-Y) stages, the Movement Disorders Society Unified Parkinson’s Disease Rating Scale (MDS-UPDRS), the Montreal Cognitive Assessment (MOCA), the Geriatric Depression Scale (GDS), the Scale for Outcomes for Parkinson’s Disease–autonomic function (SCOPA-AUT), the State and Trait Anxiety Scale (STAI), the Questionnaire for Impulsive-Compulsive Disorders in Parkinson’s Disease (QUIP), the University of Pennsylvania Smell Identification Test (UPSIT), and the Epworth Sleepiness Scale (ESS).

Striatal DAT processing

According to the PPMI imaging protocol, the baseline DAT images were acquired using [123I] FP-CIT single-photon emission computed tomography (SPECT) imaging at PPMI imaging centers. All PD patients were drug-naïve at baseline. Images were acquired using a gamma camera that is capable of performing SPECT imaging. SPECT scans were begun 4 h (± 30 min) post-radiotracer injection. The participant’s head was placed in the appropriate head holder and the field of view (FOV) encompassed the entire head to include the brain from most superior cortical regions through the inferior portion of the cerebellum. The participant was told of the necessity to avoid movements of the head and asked for her/his active cooperation. After the scan, SPECT data were sent to the Institute for Neurodegenerative Disorders for standardized processing. Striatal regions of interest (ROIs) were then placed on the target regions (caudate and putamen) and reference region (occipital cortex). Then, count densities for each striatal ROI were extracted and applied to calculate striatal binding ratios (SBR) for each striatal ROI. SBR was calculated as follows: SBR = (target region/reference region) − 1. Finally, the mean SBR of caudate and putamen was calculated. Detailed DAT processing procedures are available on the PPMI website (https://​www.​ppmiinfo.​org/​access-data-specimens/​download-data/​) and in published documents [18, 19].

Statistical analysis

To assess the demographic, clinical, and DAT imaging characteristics between the subgroups, one-way analysis of variance (ANOVA) or t-test was used to compare continuous variables that did follow a normal distribution, the Kruskal–Wallis test was used to compare continuous variables that did not follow a normal distribution, and the χ2 test and Fisher’s exact test were used for categorical variables. The Kolmogorov–Smirnov test was used to assess the normality of the continuous variables. To determine whether there was a relationship between coffee cups consumed per day and striatal DAT characteristics, partial correlation coefficients were calculated for each investigated striatal region after adjusting for age and gender in the subgroups of current and former consumers, respectively. Furthermore, we included the factors that may have influenced the loss of nigrostriatal dopaminergic neurons in multiple linear regression analyses to identify significant contributing factors to DAT availability in the investigated striatal regions. This analysis was constructed using two models. In model 1, the independent variables included age, gender, coffee consumption (current vs. former/never consumers), and smoking history (current vs. former/never smokers). Model 2 included the independent variables from model 1 as well as age at onset and disease duration that were known to be associated with dopaminergic density in PD patients. Bonferroni correction was made to adjust for multiple comparisons of striatal DAT measurements. All statistical analyses were performed using the SPSS Statistics 26 software (IBM Corporation, New York). The significance threshold was set at p < 0.05.

Results

Demographic and clinical characteristics across subgroups of coffee consumption

Demographic and clinical characteristics of the three subgroups are summarized in Table 1. A total of 138 PD patients (47 women, 34.1%) and 75 HC (24 women, 32.0%) were enrolled. There were no significant differences in age and gender among the three subgroups of PD patients and HC. However, education years were lower in PD-NC than in PD-FC (p = 0.046). In addition, MDS-UPDRS, H-Y stages, MOCA, GDS, SCOPA-AUT, STAI, QUIP, UPSIT, and ESS were also compared across the PD or HC subgroups, and these clinical characteristics showed no significant difference.
Table 1
Demographic and clinical characteristics of PD and healthy controls subgroups
 
PD-CC (n = 88)
PD-FC (n = 18)
PD-NC (n = 32)
p values
HC-CC (n = 55)
HC-FC (n = 9)
HC-NC (n = 11)
p values
Age (years)
60.1 ± 8.9
62.7 ± 10.0
57.7 ± 9.3
0.164
60.2 ± 10.4
58.7 ± 11.2
60.1 ± 11.7
0.928
Gender, ratio (F/M)
0.38 (24/64)
0.8 (8/10)
0.88 (15/17)
0.082
0.41 (16/39)
0.8 (4/5)
0.57 (4/7)
0.613
Education (years)
16.6 ± 2.5
17.4 ± 2.8
15.5 ± 2.2
0.032
17.3 ± 2.6
15.8 ± 2.0
17.1 ± 2.8
0.242
Coffee cups per day
2.0 ± 1.3
2.4 ± 2.2
NA
0.719
2.3 ± 0.9
3.1 ± 1.4
NA
0.065
Missing (number)
3
0
NA
NA
2
0
NA
NA
Current regular smokers, n (%)
1 (1%)
1 (6%)
1 (3%)
NA
0 (0%)
1 (11%)
0 (0%)
NA
Age at onset (years)
58.1 ± 9.2
61.3 ± 10.0
55.0 ± 9.5
0.068
NA
NA
NA
NA
Disease duration (months)
6.1 ± 5.5
7.5 ± 8.2
9.3 ± 9.5
0.279
NA
NA
NA
NA
H-Y stages
1.6 ± 0.5
1.6 ± 0.5
1.5 ± 0.5
0.765
NA
NA
NA
NA
MDS-UPDRS Part I
5.0 ± 3.6
4.9 ± 2.8
4.0 ± 2.7
0.442
2.9 ± 2.3
3.8 ± 3.2
3.0 ± 2.4
0.785
MDS-UPDRS Part II
5.8 ± 4.3
6.7 ± 4.2
5.2 ± 4.0
0.386
0.3 ± 0.8
0.1 ± 0.3
0.3 ± 0.5
0.657
MDS-UPDRS Part III
20.1 ± 7.5
21.8 ± 8.3
19.4 ± 9.2
0.564
0.9 ± 1.3
1.2 ± 1.1
0.6 ± 1.5
0.203
MDS-UPDRS Total Score
30.1 ± 11.6
33.4 ± 12.2
28.6 ± 12.3
0.376
4.1 ± 3.3
5.1 ± 4.0
3.9 ± 2.5
0.82
MOCA
27.3 ± 1.9
26.9 ± 2.4
27.8 ± 2.5
0.189
27.9 ± 1.0
28.4 ± 1.3
28.6 ± 1.3
0.217
GDS
1.9 ± 1.9
1.9 ± 1.5
1.9 ± 2.2
0.787
1.1 ± 1.7
1.0 ± 1.9
1.0 ± 1.2
0.73
SCOPA-AUT
8.6 ± 5.3
9.1 ± 5.0
6.8 ± 4.6
0.079
5.0 ± 2.9
5.8 ± 2.5
6.0 ± 3.6
0.496
STAI
61.7 ± 17.0
56.8 ± 17.6
61.6 ± 15.8
0.336
53.5 ± 11.7
61.7 ± 19.4
52.6 ± 13.8
0.435
QUIP
0.3 ± 0.6
0.4 ± 0.7
0.1 ± 0.4
0.238
0.4 ± 0.8
0.1 ± 0.3
0 ± 0
0.097
UPSIT
22.1 ± 7.8
24.6 ± 7.6
25.1 ± 8.0
0.151
34.7 ± 4.7
33.9 ± 5.9
33.6 ± 4.9
0.79
ESS
5.6 ± 3.3
6.0 ± 3.9
5.7 ± 3.3
0.968
5.4 ± 3.2
5.7 ± 3.9
6.3 ± 3.6
0.836
Three PD-CC did not know/prefer not to answer the question “cups per day.” Two HC-CC did not know/prefer not to answer the question “cups per day”
PD-CC current consumers of Parkinson’s Disease, PD-FC former consumers of Parkinson’s Disease, PD-NC never consumers of Parkinson’s Disease, HC-CC current consumers of healthy controls, HC-FC former consumers of healthy controls, HC-NC never consumers of healthy controls, NA not applicable, H-Y stages Hoehn and Yahr stages, MDS-UPDRS Movement Disorders Society Unified Parkinson’s Disease Rating Scale, MOCA Montreal Cognitive Assessment, GDS Geriatric Depression Scale, SCOPA-AUT Scale for Outcomes for Parkinson’s Disease—autonomic function, STAI State trait anxiety score, QUIP Questionnaire for Impulsive-Compulsive Disorders, UPSIT University of Pennsylvania Smell Identification Test, ESS Epworth Sleepiness Scale Score

Striatal DAT availability across subgroups of coffee consumption

PD patients had lower DAT availability in each striatal region compared to HC (p < 0.001) (Fig. 2, Additional file 1: Table S1). ANOVA analyses showed current coffee consumers had a tendency of lower DAT availability in the caudate than former/never coffee consumers in both PD patients and HC (Fig. 2, Table 2). In PD patients, there were significant differences in DAT availability in the caudate (p = 0.008, Bonferroni corrected) across three PD subgroups. Specifically, post hoc tests showed that current coffee consumers had significantly lower DAT availability in the caudate than former coffee consumers (p = 0.01) and never coffee consumers (p = 0.022). In HC, there were significant differences in DAT availability in the caudate (p = 0.031, Bonferroni uncorrected) across three HC subgroups. Specifically, post hoc tests showed that current coffee consumers had significantly lower DAT availability in the caudate than former coffee consumers (p = 0.022). The correlation analysis revealed that cups per day were negatively correlated with DAT availability in the caudate (r =  − 0.219, p = 0.047) in the PD-CC subgroup (Fig. 3, Additional file 1: Table S2).
Table 2
Dopamine transporter availability between subgroups of PD patients and healthy controls, respectively
 
PD-CC (n = 88)
PD-FC (n = 18)
PD-NC (n = 32)
p values
HC-CC (n = 55)
HC-FC (n = 9)
HC-NC (n = 11)
p values
Caudate
1.95 ± 0.48
2.28 ± 0.63
2.19 ± 0.41
0.008**
2.99 ± 0.59
3.52 ± 0.69
3.34 ± 0.80
0.031*
Putamen
0.79 ± 0.27
0.84 ± 0.27
0.87 ± 0.22
0.317
2.17 ± 0.55
2.34 ± 0.51
2.59 ± 0.72
0.075
Bold values indicate significant differences
PD-CC current consumers of Parkinson’s disease, PD-FC former consumers of Parkinson’s Disease, PD-NC never consumers of Parkinson’s Disease, HC-CC current consumers of healthy controls, HC-FC former consumers of healthy controls, HC-NC never consumers of healthy controls
*Bonferroni uncorrected (p < 0.05)
**Bonferroni corrected (p < 0.05/4 = 0.0125)

Independent contribution of coffee consumption to striatal DAT availability

The results of striatal DAT availability in multiple linear regression analyses of model 1 and model 2 are presented in Tables 3 and 4, respectively. Model 1 analysis showed negative associations between current coffee consumption and DAT availability in the caudate in PD patients (p = 0.004, Bonferroni corrected) and in HC (p = 0.011, Bonferroni corrected) (Table 3). In model 2, which was a stricter analysis that incorporated additional independent variables in PD patients including age at onset and disease duration, current coffee consumption remained independently and significantly negatively associated with DAT availability in the caudate (p = 0.003, Bonferroni corrected) (Table 4).
Table 3
Factors associated with dopamine transporter availability in each striatal subregion by a multiple linear regression analysis in PD patients and healthy controls
Striatal regions
Variables
Model 1 in PD (n = 138)
Model 1 in HC (n = 75)
B (SE)
95%CI
p
B (SE)
95%CI
p
Caudate
Age
 − 0.009 (0.332)
(− 0.018, 0.000)
0.060
 − 0.012 (0.007)
(− 0.026, 0.002)
0.097
Gender
0.085 (0.005)
(− 0.096, 0.265)
0.355
0.304 (0.160)
(− 0.014, 0.622)
0.061
Coffee consumption
 − 0.258 (0.087)
(− 0.430, − 0.086)
0.004
 − 0.423 (0.161)
(− 0.745, − 0.101)
0.011
Smoking history
 − 0.410 (0.289)
(− 0.982, 0.161)
0.158
 − 0.600 (0.638)
(− 1.873, 0.672)
0.350
Putamen
Age
 − 0.003 (0.002)
(− 0.008, 0.002)
0.256
 − 0.015 (0.007)
(− 0.028, − 0.002)
0.029
Gender
0.029 (0.049)
(− 0.068, 0.126)
0.553
0.154 (0.146)
(− 0.137, 0.445)
0.295
Coffee consumption
 − 0.064 (0.047)
(− 0.156, 0.029)
0.174
 − 0.319 (0.148)
(− 0.614, − 0.024)
0.035
Smoking history
 − 0.149 (0.155)
(− 0.456, 0.158)
0.339
 − 0.616 (0.584)
(− 1.782, 0.549)
0.295
Bold values indicate significant differences (Bonferroni corrected, p < 0.05/4 = 0.0125)
PD Parkinson’s disease, HC healthy controls, B unstandardized B, SE standard error, CI confidence interval
Table 4
Factors associated with dopamine transporter availability in each striatal subregion by a multiple linear regression analysis in PD patients
Striatal regions
Variables
Model 2 in PD (n = 138)
B (SE)
95%CI
p
Caudate
Age
 − 0.039 (0.026)
(− 0.089, 0.012)
0.136
Gender
0.085 (0.094)
(− 0.100, 0.270)
0.366
Coffee consumption
 − 0.262 (0.088)
(− 0.435, − 0.088)
0.003
Smoking history
 − 0.402 (0.292)
(− 0.979, 0.175)
0.170
Age at onset
0.029 (0.025)
(− 0.020, 0.078)
0.238
Disease duration
0.001 (0.006)
(− 0.011, 0.014)
0.829
Putamen
Age
 − 0.014 (0.014)
(− 0.047, 0.022)
0.326
Gender
0.034 (0.050)
(− 0.007, 0.243)
0.505
Coffee consumption
 − 0.067 (0.047)
(− 0.187, 0.047)
0.156
Smoking history
 − 0.139 (0.157)
(− 0.332, 0.446)
0.379
Age at onset
0.011 (0.013)
(− 0.022, 0.044)
0.421
Disease duration
 − 0.001 (0.003)
(− 0.014, 0.003)
0.818
Bold values indicate significant differences (Bonferroni corrected, p < 0.05/2 = 0.025)
PD Parkinson’s disease, B unstandardized B, SE standard error, CI confidence interval

Discussion

This study demonstrated that current coffee consumers had lower DAT availability in the caudate in PD patients and HC. Moreover, correlation analysis revealed that cups per day were negatively correlated with DAT availability in the caudate in PD-CC. In addition, after including the factors that may have influenced the loss of nigrostriatal dopaminergic neurons in multiple linear regression analyses, current coffee consumption remained an independent predictor of decreased DAT availability in the caudate in both PD patients and HC.
Caffeine antagonizes adenosine receptors (i.e., A1, A2A, A3, and A2B), contributing to hyperexcitability of the central nervous system [1, 20]. A neuroprotective effect of caffeine is well documented in experimental PD models and is probably mediated by antagonizing adenosine receptors [21, 22]. Several experiments showed that caffeine pretreatment before 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration resisted dopamine depletion in a dose-dependent manner in mice [23, 24]. In MPTP mice, the residual dopamine level was 40% of control values with caffeine pretreatment (10 mg/kg), whereas it was 15% of control values without caffeine pretreatment [24]. At 20 mg/kg, caffeine pretreatment nearly reversed the dopamine depletion produced by MPTP in mice [24]. Furthermore, another experiment demonstrated that delayed caffeine administration could also reduce the loss of nigral dopamine cell bodies and block the nigral neurodegenerative process in rats [25]. DAT is expressed in the dopaminergic neurons and mediates the reuptake of free dopamine from the synaptic cleft back into the axonal button, thereby regulating dopamine levels [26]. This study demonstrated that current coffee consumers had lower DAT availability in the caudate in PD (p = 0.008, Bonferroni corrected) and HC (p = 0.031, Bonferroni uncorrected). In HC, the p-value (p = 0.031) did not pass the strict multiple comparison correction (Bonferroni correction). We speculate that this may be due to the small sample size of the HC group compared to the PD group (75 vs. 138 participants). Further multiple linear regression analyses supported this result that current coffee consumption was negatively associated with DAT availability in the caudate compared with former/never consumers in the HC group (similar to the results in the PD group). Caffeine releases the tonic inhibition of dopamine, boosts dopamine release, and induces a secondary reduced DAT availability [16, 27]. In PD, the loss of dopaminergic neurons caused by nigrostriatal degeneration results in a substantial reduction of the DAT density and dopamine levels [28]. Despite the similar reduced changes in striatal DAT availability caused by PD pathologies and caffeine, the inverse alteration of dopamine levels caused by PD pathologies and caffeine may provide the reason for the favorable implication of caffeine to PD pathologies (Fig. 4). Gigante et al. investigated the association between chronic coffee consumption and striatal DAT binding in PD patients, including 71 current coffee consumers and 12 never consumers. However, they reported negative results [29]. We believe that the discrepancies in the results may be related to the differences in sample size (83 vs. 138 PD participants), coffee consumption categorization (two categories vs. three categories), and analytical methods (caudate-based vs. alternative anatomical analyses). Furthermore, the present study is the first to detect a significant decrease in striatal DAT availability only in the current consumers but not the former consumers compared with never consumers, which suggests that effects of caffeine on striatal DAT availability may fade and disappear after quitting coffee consumption.
In addition, this study demonstrated that current coffee consumption was significantly negatively associated with DAT availability in the caudate but not in the putamen compared with former/never consumers in PD patients and HC. This finding indicates that caudate is more susceptible to the effects of coffee consumption compared with putamen. Using in vivo microdialysis, previous studies found caffeine increased striatal extracellular dopamine levels by antagonizing the adenosine A1 receptor [30, 31]. A molecular imaging study found that human caudate showed relative enrichment for adenosine A1 receptor compared to putamen (88 ± 4 vs. 83 ± 4) [32], which appears to be the main driver of caffeine facilitative effect on dopamine release [30]. In addition, caudate dopaminergic dysfunction is commonly seen in PD patients and plays a crucial role in the pathophysiology of parkinsonian symptoms such as cognitive impairment [33], depression [34], rapid eye movement sleep behavior disorder [35], and gait problems [36]. In addition, baseline caudate dopaminergic dysfunction is associated with an increased risk of developing cognitive impairment, depression, and gait problems over the follow-up [37]. At present, the clinical use of caffeine is not considered a contraindication to or even a confound of DAT imaging. Although current coffee consumption may cause a decrease in DAT availability, the descending range in DAT availability caused by caffeine is much lower than that caused by PD (Fig. 2). However, we believe that caffeine may lead to a false-positive diagnosis of some diseases for which the DAT availability decline is not yet obvious, such as prodromal PD. Therefore, whether coffee use should be banned before scanning is a question that needs further exploration, especially for some suspected diseases with decreased DAT availability in the caudate.
Although the present study showed chronic coffee consumption was associated with decreased striatal DAT availability, the retrospective study design in this study does not allow us to determine causality; we cannot determine whether the alterations of DAT availability predispose to coffee consumption initiation or whether chronic coffee consumption influences DAT availability. A case–control study investigated the association between cigarette smoking, alcohol drinking, coffee consumption, and PD risk [38]. The results showed coffee consumption was the highest factor in the reduction of PD risk. This strong inverse relationship remained stable when ever versus never coffee consumption was considered (OR = 0.16, p = 0.0001). This indicates that the proportion of never coffee consumers is higher in PD than in HC in that epidemiology study [38]. In the present study, the proportion of never coffee consumers in the PD group appears to be relatively higher than HC (23.2% vs. 14.7%). We compared the proportion of never coffee consumers between PD and HC using the chi-square test. However, there were no significant differences in the proportion of never coffee consumers between PD and HC (p = 0.139). It is still unclear whether predisposing factors, such as the genetic factors that make it easy to get into the habit of drinking coffee, the effects of chronic coffee consumption that lead to DAT availability abnormalities, or a combination of factors, are involved. Prospective longitudinal studies could answer this question. In the future, longitudinal imaging investigations should be designed to explore DAT availability changes before and after the initiation of coffee consumption.
In this study, the multiple linear regression analysis included smoking history (current vs. former/never smokers) as an independent variable. Unexpectedly, current smoking did not show any association with striatal DAT availability. Like coffee, cigarette smoking is a well-established protective factor for PD [39]. An inverse association between cigarette smoking and the risk of PD is well documented [40]. Several studies have found decreased striatal DAT availability in current smokers compared to non-smokers [4143]. Nevertheless, after the onset, PD patients are able to quit smoking more easily than controls [44]. We believe that no significant results in our study may be mainly related to the small sample size of current smokers. In our study, only three PD patients were current smokers, and only one control was a current smoker.
The main strength of the present study is the large sample that allows testing different (current/former/never) coffee consumption effects on striatal DAT availability in both PD patients and HC. This study has several limitations. First, although several known confounding factors that significantly influence dopaminergic density were included in this analysis, it is possible that other lifestyle-related confounding factors may have altered the results. Second, similar to coffee, smoking is a well-established protective factor for PD [39]. However, PD patients are able to quit smoking more easily than healthy controls [44]. In this study, no significant association between current smoking and DAT may mainly be related to the small sample size of current smokers (only 3 PD patients). Third, although SPECT scans were begun 4 h (± 30 min) post-radiotracer injection according to the PPMI imaging protocol, information regarding the time of the last coffee cup consumed before SPECT scans was not available. Future studies should investigate the acute effects of coffee consumption on striatal DAT availability. Fourth, we investigated the association between caffeinated coffee consumption and striatal DAT availability in this study. Therefore, the findings of this study may not apply to decaffeinated coffee consumers. In addition to caffeine, there are many other potentially central nervous system active compounds in coffee that may alter DAT availability. Future studies should explore whether decaffeinated coffee consumption influences striatal DAT availability.

Conclusions

In summary, this study demonstrated that current coffee consumption was associated with decreased striatal DAT availability in the caudate. Caffeine acts as a stimulant to boost dopamine release and induce a secondary reduced DAT availability, while the degeneration of dopaminergic neurons results in a substantial reduction of the DAT density and dopamine levels in PD. The inverse change of striatal dopamine levels caused by caffeine and PD pathologies may explain the favorable implication of caffeine to PD pathologies. Our study provides evidence for the favorable implication of caffeine to PD pathologies using human brain imaging in vivo. In addition, our study showed that reduced striatal DAT availability was detected in current coffee consumers but not in former coffee consumers compared with never coffee consumers, suggesting that caffeine’s effects on striatal DAT may fade and disappear after quitting coffee consumption.

Acknowledgements

Data used in the preparation of this article were obtained from the Parkinson’s Progression Markers Initiative (PPMI) database (http://​www.​ppmi-info.​org/​access-data-specimens/​download-data). For up-to-date information on the study, visit http://​www.​ppmi-info.​org. PPMI—a public-private partnership—is funded by the Michael J. Fox Foundation for Parkinson’s Research and funding partners, including AbbVie, Allergan, Avid, Biogen, Bristol-Myers Squibb, Covance, GE Healthcare, Genentech, GlaxoSmithKline, Lilly, Lundbeck, Merck, Meso Scale Discovery, Pfizer, Piramal, Roche, SANOFI GENZYME, Servier, Takeda, Teva, UCB, and GolubCapital.

Declarations

The PPMI project was approved by the Institutional Review Board or Independent Ethics Committee of all participating sites in Europe, including Attikon University Hospital (Greece), Hospital Clinic de Barcelona and Hospital Universitario Donostia (Spain), Innsbruck University (Austria), Paracelsus-Elena-Klinic Kassel/University of Marburg (Germany), Imperial College London (UK), Pitié-Salpêtrière Hospital (France), and University of Salerno (Italy), and in the USA, including Emory University, Johns Hopkins University, University of Alabama at Birmingham, PD and Movement Disorders Center of Boca Raton, Boston University, Northwestern University, University of Cincinnati, Cleveland Clinic Foundation, Baylor College of Medicine, Institute for Neurodegenerative Disorders, Columbia University Medical Center, Beth Israel Medical Center, University of Pennsylvania, Oregon Health and Science University, University of Rochester, University of California at San Diego, and University of California, San Francisco. Detailed information is available at http://​ppmi-info.​org/​ppmiclinical-sites. The relevant local institutional review boards approved the PPMI protocol and written informed consent was obtained from all participants prior to inclusion. No additional ethics approval was required from the local ethics committee where data was analyzed.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Fredholm BB, Bättig K, Holmén J, Nehlig A, Zvartau EE. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev. 1999;51(1):83–133.PubMed Fredholm BB, Bättig K, Holmén J, Nehlig A, Zvartau EE. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev. 1999;51(1):83–133.PubMed
2.
Zurück zum Zitat Ullrich S, de Vries YC, Kühn S, Repantis D, Dresler M, Ohla K. Feeling smart: effects of caffeine and glucose on cognition, mood and self-judgment. Physiol Behav. 2015;151:629–37.PubMedCrossRef Ullrich S, de Vries YC, Kühn S, Repantis D, Dresler M, Ohla K. Feeling smart: effects of caffeine and glucose on cognition, mood and self-judgment. Physiol Behav. 2015;151:629–37.PubMedCrossRef
3.
4.
Zurück zum Zitat O’Keefe JH, DiNicolantonio JJ, Lavie CJ. Coffee for cardioprotection and longevity. Prog Cardiovasc Dis. 2018;61(1):38–42.PubMedCrossRef O’Keefe JH, DiNicolantonio JJ, Lavie CJ. Coffee for cardioprotection and longevity. Prog Cardiovasc Dis. 2018;61(1):38–42.PubMedCrossRef
5.
Zurück zum Zitat Ross GW, Abbott RD, Petrovitch H, Morens DM, Grandinetti A, Tung KH, Tanner CM, Masaki KH, Blanchette PL, Curb JD, et al. Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA. 2000;283(20):2674–9.PubMedCrossRef Ross GW, Abbott RD, Petrovitch H, Morens DM, Grandinetti A, Tung KH, Tanner CM, Masaki KH, Blanchette PL, Curb JD, et al. Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA. 2000;283(20):2674–9.PubMedCrossRef
6.
Zurück zum Zitat Sääksjärvi K, Knekt P, Rissanen H, Laaksonen MA, Reunanen A, Männistö S. Prospective study of coffee consumption and risk of Parkinson’s disease. Eur J Clin Nutr. 2008;62(7):908–15.PubMedCrossRef Sääksjärvi K, Knekt P, Rissanen H, Laaksonen MA, Reunanen A, Männistö S. Prospective study of coffee consumption and risk of Parkinson’s disease. Eur J Clin Nutr. 2008;62(7):908–15.PubMedCrossRef
7.
Zurück zum Zitat Qi H, Li S. Dose-response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson’s disease. Geriatr Gerontol Int. 2014;14(2):430–9.PubMedCrossRef Qi H, Li S. Dose-response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson’s disease. Geriatr Gerontol Int. 2014;14(2):430–9.PubMedCrossRef
8.
Zurück zum Zitat Palacios N, Gao X, McCullough ML, Schwarzschild MA, Shah R, Gapstur S, Ascherio A. Caffeine and risk of Parkinson’s disease in a large cohort of men and women. Mov Disord. 2012;27(10):1276–82.PubMedPubMedCentralCrossRef Palacios N, Gao X, McCullough ML, Schwarzschild MA, Shah R, Gapstur S, Ascherio A. Caffeine and risk of Parkinson’s disease in a large cohort of men and women. Mov Disord. 2012;27(10):1276–82.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.PubMedCrossRef Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.PubMedCrossRef
10.
Zurück zum Zitat Dickson DW, Braak H, Duda JE, Duyckaerts C, Gasser T, Halliday GM, Hardy J, Leverenz JB, Del Tredici K, Wszolek ZK, et al. Neuropathological assessment of Parkinson’s disease: refining the diagnostic criteria. Lancet Neurol. 2009;8(12):1150–7.PubMedCrossRef Dickson DW, Braak H, Duda JE, Duyckaerts C, Gasser T, Halliday GM, Hardy J, Leverenz JB, Del Tredici K, Wszolek ZK, et al. Neuropathological assessment of Parkinson’s disease: refining the diagnostic criteria. Lancet Neurol. 2009;8(12):1150–7.PubMedCrossRef
11.
Zurück zum Zitat Ojeda-López C, Cervantes-Arriaga A, Rodríguez-Violante M, Corona T. Caffeine drinking, cigarette smoking, and dopaminergic replacement therapy dose in Parkinson’s disease. Neurol Sci. 2013;34(6):979–83.PubMedCrossRef Ojeda-López C, Cervantes-Arriaga A, Rodríguez-Violante M, Corona T. Caffeine drinking, cigarette smoking, and dopaminergic replacement therapy dose in Parkinson’s disease. Neurol Sci. 2013;34(6):979–83.PubMedCrossRef
12.
Zurück zum Zitat Nicoletti A, Zappia M. Coffee consumption and risk of levodopa-induced dyskinesia in Parkinson’s disease: the FRAGAMP study. Mov Disord. 2015;30(13):1854–6.PubMedCrossRef Nicoletti A, Zappia M. Coffee consumption and risk of levodopa-induced dyskinesia in Parkinson’s disease: the FRAGAMP study. Mov Disord. 2015;30(13):1854–6.PubMedCrossRef
13.
Zurück zum Zitat Magalhães R, Picó-Pérez M, Esteves M, Vieira R, Castanho TC, Amorim L, Sousa M, Coelho A, Fernandes HM, Cabral J, et al. Habitual coffee drinkers display a distinct pattern of brain functional connectivity. Mol Psychiatry. 2021;26(11):6589–98.PubMedPubMedCentralCrossRef Magalhães R, Picó-Pérez M, Esteves M, Vieira R, Castanho TC, Amorim L, Sousa M, Coelho A, Fernandes HM, Cabral J, et al. Habitual coffee drinkers display a distinct pattern of brain functional connectivity. Mol Psychiatry. 2021;26(11):6589–98.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Giros B, Caron MG. Molecular characterization of the dopamine transporter. Trends Pharmacol Sci. 1993;14(2):43–9.PubMedCrossRef Giros B, Caron MG. Molecular characterization of the dopamine transporter. Trends Pharmacol Sci. 1993;14(2):43–9.PubMedCrossRef
15.
Zurück zum Zitat Uhl GR. Dopamine transporter: basic science and human variation of a key molecule for dopaminergic function, locomotion, and parkinsonism. Mov Disord. 2003;18(Suppl 7):S71-80.PubMedCrossRef Uhl GR. Dopamine transporter: basic science and human variation of a key molecule for dopaminergic function, locomotion, and parkinsonism. Mov Disord. 2003;18(Suppl 7):S71-80.PubMedCrossRef
16.
Zurück zum Zitat Petzer JP, Petzer A. Caffeine as a lead compound for the design of therapeutic agents for the treatment of Parkinson’s disease. Curr Med Chem. 2015;22(8):975–88.PubMedCrossRef Petzer JP, Petzer A. Caffeine as a lead compound for the design of therapeutic agents for the treatment of Parkinson’s disease. Curr Med Chem. 2015;22(8):975–88.PubMedCrossRef
17.
Zurück zum Zitat Schwarzschild MA, Xu K, Oztas E, Petzer JP, Castagnoli K, Castagnoli N Jr, Chen JF. Neuroprotection by caffeine and more specific A2A receptor antagonists in animal models of Parkinson’s disease. Neurology. 2003;61(11 Suppl 6):S55-61.PubMedCrossRef Schwarzschild MA, Xu K, Oztas E, Petzer JP, Castagnoli K, Castagnoli N Jr, Chen JF. Neuroprotection by caffeine and more specific A2A receptor antagonists in animal models of Parkinson’s disease. Neurology. 2003;61(11 Suppl 6):S55-61.PubMedCrossRef
18.
Zurück zum Zitat Tinaz S, Chow C, Kuo PH, Krupinski EA, Blumenfeld H, Louis ED, Zubal G. Semiquantitative analysis of dopamine transporter scans in patients with Parkinson disease. Clin Nucl Med. 2018;43(1):e1–7.PubMedPubMedCentralCrossRef Tinaz S, Chow C, Kuo PH, Krupinski EA, Blumenfeld H, Louis ED, Zubal G. Semiquantitative analysis of dopamine transporter scans in patients with Parkinson disease. Clin Nucl Med. 2018;43(1):e1–7.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Tagare HD, DeLorenzo C, Chelikani S, Saperstein L, Fulbright RK. Voxel-based logistic analysis of PPMI control and Parkinson’s disease DaTscans. Neuroimage. 2017;152:299–311.PubMedCrossRef Tagare HD, DeLorenzo C, Chelikani S, Saperstein L, Fulbright RK. Voxel-based logistic analysis of PPMI control and Parkinson’s disease DaTscans. Neuroimage. 2017;152:299–311.PubMedCrossRef
20.
21.
Zurück zum Zitat Kachroo A, Irizarry MC, Schwarzschild MA. Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration. Exp Neurol. 2010;223(2):657–61.PubMedPubMedCentralCrossRef Kachroo A, Irizarry MC, Schwarzschild MA. Caffeine protects against combined paraquat and maneb-induced dopaminergic neuron degeneration. Exp Neurol. 2010;223(2):657–61.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Xu K, Xu YH, Chen JF, Schwarzschild MA. Neuroprotection by caffeine: time course and role of its metabolites in the MPTP model of Parkinson’s disease. Neuroscience. 2010;167(2):475–81.PubMedCrossRef Xu K, Xu YH, Chen JF, Schwarzschild MA. Neuroprotection by caffeine: time course and role of its metabolites in the MPTP model of Parkinson’s disease. Neuroscience. 2010;167(2):475–81.PubMedCrossRef
23.
Zurück zum Zitat Xu K, Xu Y, Brown-Jermyn D, Chen JF, Ascherio A, Dluzen DE, Schwarzschild MA. Estrogen prevents neuroprotection by caffeine in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci. 2006;26(2):535–41.PubMedPubMedCentralCrossRef Xu K, Xu Y, Brown-Jermyn D, Chen JF, Ascherio A, Dluzen DE, Schwarzschild MA. Estrogen prevents neuroprotection by caffeine in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson’s disease. J Neurosci. 2006;26(2):535–41.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Chen JF, Xu K, Petzer JP, Staal R, Xu YH, Beilstein M, Sonsalla PK, Castagnoli K, Castagnoli N, Jr., Schwarzschild MA. Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson’s disease. J Neurosci. 2001;21(10):Rc143. Chen JF, Xu K, Petzer JP, Staal R, Xu YH, Beilstein M, Sonsalla PK, Castagnoli K, Castagnoli N, Jr., Schwarzschild MA. Neuroprotection by caffeine and A(2A) adenosine receptor inactivation in a model of Parkinson’s disease. J Neurosci. 2001;21(10):Rc143.
25.
Zurück zum Zitat Sonsalla PK, Wong LY, Harris SL, Richardson JR, Khobahy I, Li W, Gadad BS, German DC. Delayed caffeine treatment prevents nigral dopamine neuron loss in a progressive rat model of Parkinson’s disease. Exp Neurol. 2012;234(2):482–7.PubMedPubMedCentralCrossRef Sonsalla PK, Wong LY, Harris SL, Richardson JR, Khobahy I, Li W, Gadad BS, German DC. Delayed caffeine treatment prevents nigral dopamine neuron loss in a progressive rat model of Parkinson’s disease. Exp Neurol. 2012;234(2):482–7.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Kurosaki R, Muramatsu Y, Watanabe H, Michimata M, Matsubara M, Imai Y, Araki T. Role of dopamine transporter against MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) neurotoxicity in mice. Metab Brain Dis. 2003;18(2):139–46.PubMedCrossRef Kurosaki R, Muramatsu Y, Watanabe H, Michimata M, Matsubara M, Imai Y, Araki T. Role of dopamine transporter against MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) neurotoxicity in mice. Metab Brain Dis. 2003;18(2):139–46.PubMedCrossRef
27.
Zurück zum Zitat Dunwiddie TV, Masino SA. The role and regulation of adenosine in the central nervous system. Annu Rev Neurosci. 2001;24:31–55.PubMedCrossRef Dunwiddie TV, Masino SA. The role and regulation of adenosine in the central nervous system. Annu Rev Neurosci. 2001;24:31–55.PubMedCrossRef
28.
Zurück zum Zitat Kish SJ, Shannak K, Hornykiewicz O. Uneven pattern of dopamine loss in the striatum of patients with idiopathic Parkinson’s disease. Pathophysiologic and clinical implications. N Engl J Med. 1988;318(14):876–880. Kish SJ, Shannak K, Hornykiewicz O. Uneven pattern of dopamine loss in the striatum of patients with idiopathic Parkinson’s disease. Pathophysiologic and clinical implications. N Engl J Med. 1988;318(14):876–880.
29.
Zurück zum Zitat Gigante AF, Asabella AN, Iliceto G, Martino T, Ferrari C, Defazio G, Rubini G. Chronic coffee consumption and striatal DAT-SPECT findings in Parkinson’s disease. Neurol Sci. 2018;39(3):551–5.PubMedCrossRef Gigante AF, Asabella AN, Iliceto G, Martino T, Ferrari C, Defazio G, Rubini G. Chronic coffee consumption and striatal DAT-SPECT findings in Parkinson’s disease. Neurol Sci. 2018;39(3):551–5.PubMedCrossRef
30.
Zurück zum Zitat Borycz J, Pereira MF, Melani A, Rodrigues RJ, Köfalvi A, Panlilio L, Pedata F, Goldberg SR, Cunha RA, Ferré S. Differential glutamate-dependent and glutamate-independent adenosine A1 receptor-mediated modulation of dopamine release in different striatal compartments. J Neurochem. 2007;101(2):355–63.PubMedCrossRef Borycz J, Pereira MF, Melani A, Rodrigues RJ, Köfalvi A, Panlilio L, Pedata F, Goldberg SR, Cunha RA, Ferré S. Differential glutamate-dependent and glutamate-independent adenosine A1 receptor-mediated modulation of dopamine release in different striatal compartments. J Neurochem. 2007;101(2):355–63.PubMedCrossRef
31.
Zurück zum Zitat Okada M, Kiryu K, Kawata Y, Mizuno K, Wada K, Tasaki H, Kaneko S. Determination of the effects of caffeine and carbamazepine on striatal dopamine release by in vivo microdialysis. Eur J Pharmacol. 1997;321(2):181–8.PubMedCrossRef Okada M, Kiryu K, Kawata Y, Mizuno K, Wada K, Tasaki H, Kaneko S. Determination of the effects of caffeine and carbamazepine on striatal dopamine release by in vivo microdialysis. Eur J Pharmacol. 1997;321(2):181–8.PubMedCrossRef
32.
Zurück zum Zitat Ikeda M, Mackay KB, Dewar D, McCulloch J. Differential alterations in adenosine A1 and kappa 1 opioid receptors in the striatum in Alzheimer’s disease. Brain Res. 1993;616(1–2):211–7.PubMedCrossRef Ikeda M, Mackay KB, Dewar D, McCulloch J. Differential alterations in adenosine A1 and kappa 1 opioid receptors in the striatum in Alzheimer’s disease. Brain Res. 1993;616(1–2):211–7.PubMedCrossRef
33.
Zurück zum Zitat Schrag A, Siddiqui UF, Anastasiou Z, Weintraub D, Schott JM. Clinical variables and biomarkers in prediction of cognitive impairment in patients with newly diagnosed Parkinson’s disease: a cohort study. Lancet Neurol. 2017;16(1):66–75.PubMedCrossRef Schrag A, Siddiqui UF, Anastasiou Z, Weintraub D, Schott JM. Clinical variables and biomarkers in prediction of cognitive impairment in patients with newly diagnosed Parkinson’s disease: a cohort study. Lancet Neurol. 2017;16(1):66–75.PubMedCrossRef
34.
Zurück zum Zitat Vriend C, Raijmakers P, Veltman DJ, van Dijk KD, van der Werf YD, Foncke EM, Smit JH, Berendse HW, van den Heuvel OA. Depressive symptoms in Parkinson’s disease are related to reduced [123I]FP-CIT binding in the caudate nucleus. J Neurol Neurosurg Psychiatry. 2014;85(2):159–64.PubMedCrossRef Vriend C, Raijmakers P, Veltman DJ, van Dijk KD, van der Werf YD, Foncke EM, Smit JH, Berendse HW, van den Heuvel OA. Depressive symptoms in Parkinson’s disease are related to reduced [123I]FP-CIT binding in the caudate nucleus. J Neurol Neurosurg Psychiatry. 2014;85(2):159–64.PubMedCrossRef
35.
Zurück zum Zitat Arnaldi D, De Carli F, Picco A, Ferrara M, Accardo J, Bossert I, Famà F, Girtler N, Morbelli S, Sambuceti G, et al. Nigro-caudate dopaminergic deafferentation: a marker of REM sleep behavior disorder? Neurobiol Aging. 2015;36(12):3300–5.PubMedCrossRef Arnaldi D, De Carli F, Picco A, Ferrara M, Accardo J, Bossert I, Famà F, Girtler N, Morbelli S, Sambuceti G, et al. Nigro-caudate dopaminergic deafferentation: a marker of REM sleep behavior disorder? Neurobiol Aging. 2015;36(12):3300–5.PubMedCrossRef
36.
Zurück zum Zitat Ouchi Y, Kanno T, Okada H, Yoshikawa E, Futatsubashi M, Nobezawa S, Torizuka T, Tanaka K. Changes in dopamine availability in the nigrostriatal and mesocortical dopaminergic systems by gait in Parkinson’s disease. Brain. 2001;124(Pt 4):784–92.PubMedCrossRef Ouchi Y, Kanno T, Okada H, Yoshikawa E, Futatsubashi M, Nobezawa S, Torizuka T, Tanaka K. Changes in dopamine availability in the nigrostriatal and mesocortical dopaminergic systems by gait in Parkinson’s disease. Brain. 2001;124(Pt 4):784–92.PubMedCrossRef
37.
Zurück zum Zitat Pasquini J, Durcan R, Wiblin L, Gersel Stokholm M, Rochester L, Brooks DJ, Burn D, Pavese N. Clinical implications of early caudate dysfunction in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2019;90(10):1098–104.PubMedCrossRef Pasquini J, Durcan R, Wiblin L, Gersel Stokholm M, Rochester L, Brooks DJ, Burn D, Pavese N. Clinical implications of early caudate dysfunction in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2019;90(10):1098–104.PubMedCrossRef
38.
Zurück zum Zitat Ragonese P, Salemi G, Morgante L, Aridon P, Epifanio A, Buffa D, Scoppa F, Savettieri G. A case-control study on cigarette, alcohol, and coffee consumption preceding Parkinson’s disease. Neuroepidemiology. 2003;22(5):297–304.PubMedCrossRef Ragonese P, Salemi G, Morgante L, Aridon P, Epifanio A, Buffa D, Scoppa F, Savettieri G. A case-control study on cigarette, alcohol, and coffee consumption preceding Parkinson’s disease. Neuroepidemiology. 2003;22(5):297–304.PubMedCrossRef
39.
Zurück zum Zitat Gallo V, Vineis P, Cancellieri M, Chiodini P, Barker RA, Brayne C, Pearce N, Vermeulen R, Panico S, Bueno-de-Mesquita B, et al. Exploring causality of the association between smoking and Parkinson’s disease. Int J Epidemiol. 2019;48(3):912–25.PubMed Gallo V, Vineis P, Cancellieri M, Chiodini P, Barker RA, Brayne C, Pearce N, Vermeulen R, Panico S, Bueno-de-Mesquita B, et al. Exploring causality of the association between smoking and Parkinson’s disease. Int J Epidemiol. 2019;48(3):912–25.PubMed
40.
Zurück zum Zitat Mappin-Kasirer B, Pan H, Lewington S, Kizza J, Gray R, Clarke R, Peto R. Tobacco smoking and the risk of Parkinson disease: a 65-year follow-up of 30,000 male British doctors. Neurology. 2020;94(20):e2132–8.PubMedPubMedCentralCrossRef Mappin-Kasirer B, Pan H, Lewington S, Kizza J, Gray R, Clarke R, Peto R. Tobacco smoking and the risk of Parkinson disease: a 65-year follow-up of 30,000 male British doctors. Neurology. 2020;94(20):e2132–8.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Newberg A, Lerman C, Wintering N, Ploessl K, Mozley PD. Dopamine transporter binding in smokers and nonsmokers. Clin Nucl Med. 2007;32(6):452–5.PubMedCrossRef Newberg A, Lerman C, Wintering N, Ploessl K, Mozley PD. Dopamine transporter binding in smokers and nonsmokers. Clin Nucl Med. 2007;32(6):452–5.PubMedCrossRef
42.
Zurück zum Zitat Yang YK, Yao WJ, Yeh TL, Lee IH, Chen PS, Lu RB, Chiu NT. Decreased dopamine transporter availability in male smokers – a dual isotope SPECT study. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(1):274–9.PubMedCrossRef Yang YK, Yao WJ, Yeh TL, Lee IH, Chen PS, Lu RB, Chiu NT. Decreased dopamine transporter availability in male smokers – a dual isotope SPECT study. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32(1):274–9.PubMedCrossRef
43.
Zurück zum Zitat Wang C, Zhou C, Guo T, Huang P, Xu X, Zhang M. Association between cigarette smoking and Parkinson’s disease: a neuroimaging study. Ther Adv Neurol Disord. 2022;15:17562864221092566.PubMedPubMedCentralCrossRef Wang C, Zhou C, Guo T, Huang P, Xu X, Zhang M. Association between cigarette smoking and Parkinson’s disease: a neuroimaging study. Ther Adv Neurol Disord. 2022;15:17562864221092566.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Ritz B, Lee PC, Lassen CF, Arah OA. Parkinson disease and smoking revisited: ease of quitting is an early sign of the disease. Neurology. 2014;83(16):1396–402.PubMedPubMedCentralCrossRef Ritz B, Lee PC, Lassen CF, Arah OA. Parkinson disease and smoking revisited: ease of quitting is an early sign of the disease. Neurology. 2014;83(16):1396–402.PubMedPubMedCentralCrossRef
Metadaten
Titel
Current coffee consumption is associated with decreased striatal dopamine transporter availability in Parkinson’s disease patients and healthy controls
verfasst von
Chao Wang
Cheng Zhou
Tao Guo
Yeerfan Jiaerken
Siyu Yang
Xiaopei Xu
Ling Hu
Peiyu Huang
Xiaojun Xu
Minming Zhang
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2023
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-023-02994-5

Weitere Artikel der Ausgabe 1/2023

BMC Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 49 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

Wie der Klimawandel gefährliche Pilzinfektionen begünstigt

24.05.2024 Candida-Mykosen Nachrichten

Dass sich invasive Pilzinfektionen in letzter Zeit weltweit häufen, liegt wahrscheinlich auch am Klimawandel. Ausbrüche mit dem Hefepilz Candida auris stellen eine zunehmende Gefahr für Immungeschwächte dar – auch in Deutschland.

So wirken verschiedene Alkoholika auf den Blutdruck

23.05.2024 Störungen durch Alkohol Nachrichten

Je mehr Alkohol Menschen pro Woche trinken, desto mehr steigt ihr Blutdruck, legen Daten aus Dänemark nahe. Ob es dabei auch auf die Art des Alkohols ankommt, wurde ebenfalls untersucht.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

Update Allgemeinmedizin

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