Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
Add more filters

Publication year range
1.
J Clin Psychopharmacol ; 44(4): 386-396, 2024.
Article in English | MEDLINE | ID: mdl-38901008

ABSTRACT

BACKGROUND: Deutetrabenazine is approved for adults with tardive dyskinesia (TD). Data based on underlying psychiatric condition and baseline dopamine receptor antagonist (DRA) use are limited. METHODS: Patients with TD who completed parent studies ARM-TD or AIM-TD were eligible for the 3-year, open-label extension study (RIM-TD; NCT02198794). In RIM-TD, deutetrabenazine was titrated based on dyskinesia control and tolerability. In this post hoc analysis of RIM-TD, total motor Abnormal Involuntary Movement Scale (AIMS) score and adverse events (AEs) were analyzed by underlying condition and DRA use at parent study baseline. RESULTS: Of 343 patients enrolled in RIM-TD, 336 were included in the analysis by underlying condition, and 337 were included in the analysis by DRA use. One hundred eighty-nine of 205 (92%) patients with psychotic disorders (schizophrenia/schizoaffective disorder) and 65 of 131 (50%) with mood and other disorders (depression/bipolar disorder/other) were receiving a DRA. Mean (SE) deutetrabenazine doses at week 145 were 40.4 (1.13), 38.5 (1.21), 39.9 (1.00), and 38.5 (1.48) mg/d for patients with psychotic disorders, those with mood and other disorders, and those receiving DRAs or not, respectively. Mean (SD) changes in total motor AIMS score from this study baseline to week 145 were -6.3 (4.53), -7.1 (4.92), -6.1 (4.42), and -7.5 (5.19). Exposure-adjusted incidence rates (number of AEs/patient-years) of AEs were similar across groups: any (1.02, 1.71, 1.08, 1.97), serious (0.10, 0.12, 0.10, 0.12), and leading to discontinuation (0.07, 0.05, 0.06, 0.05). CONCLUSIONS: Long-term deutetrabenazine provided clinically meaningful improvements in TD-related movements, with a favorable benefit-risk profile, regardless of underlying condition or DRA use.


Subject(s)
Dopamine Antagonists , Tardive Dyskinesia , Tetrabenazine , Humans , Tardive Dyskinesia/drug therapy , Tardive Dyskinesia/chemically induced , Male , Female , Tetrabenazine/analogs & derivatives , Tetrabenazine/pharmacology , Tetrabenazine/adverse effects , Tetrabenazine/administration & dosage , Middle Aged , Adult , Dopamine Antagonists/adverse effects , Dopamine Antagonists/administration & dosage , Dopamine Antagonists/pharmacology , Psychotic Disorders/drug therapy , Aged , Antipsychotic Agents/adverse effects , Antipsychotic Agents/administration & dosage , Schizophrenia/drug therapy , Treatment Outcome
2.
Neurobiol Dis ; 179: 106048, 2023 04.
Article in English | MEDLINE | ID: mdl-36813207

ABSTRACT

BACKGROUND: Freezing of gait (FOG) is a major cause of falling in Parkinson's disease (PD) and can be responsive or unresponsive to levodopa. Pathophysiology is poorly understood. OBJECTIVE: To examine the link between noradrenergic systems, the development of FOG in PD and its responsiveness to levodopa. METHODS: We examined norepinephrine transporter (NET) binding via brain positron emission tomography (PET) to evaluate changes in NET density associated with FOG using the high affinity selective NET antagonist radioligand [11C]MeNER (2S,3S)(2-[α-(2-methoxyphenoxy)benzyl]morpholine) in 52 parkinsonian patients. We used a rigorous levodopa challenge paradigm to characterize PD patients as non-freezing (NO-FOG, N = 16), levodopa responsive freezing (OFF-FOG, N = 10), and levodopa-unresponsive freezing (ONOFF-FOG, N = 21), and also included a non-PD FOG group, primary progressive freezing of gait (PP-FOG, N = 5). RESULTS: Linear mixed models identified significant reductions in whole brain NET binding in the OFF-FOG group compared to the NO-FOG group (-16.8%, P = 0.021) and regionally in the frontal lobe, left and right thalamus, temporal lobe, and locus coeruleus, with the strongest effect in right thalamus (P = 0.038). Additional regions examined in a post hoc secondary analysis including the left and right amygdalae confirmed the contrast between OFF-FOG and NO-FOG (P = 0.003). A linear regression analysis identified an association between reduced NET binding in the right thalamus and more severe New FOG Questionnaire (N-FOG-Q) score only in the OFF-FOG group (P = 0.022). CONCLUSION: This is the first study to examine brain noradrenergic innervation using NET-PET in PD patients with and without FOG. Based on the normal regional distribution of noradrenergic innervation and pathological studies in the thalamus of PD patients, the implications of our findings suggest that noradrenergic limbic pathways may play a key role in OFF-FOG in PD. This finding could have implications for clinical subtyping of FOG as well as development of therapies.


Subject(s)
Gait Disorders, Neurologic , Parkinson Disease , Humans , Parkinson Disease/complications , Parkinson Disease/diagnostic imaging , Parkinson Disease/drug therapy , Levodopa/therapeutic use , Norepinephrine Plasma Membrane Transport Proteins , Gait Disorders, Neurologic/diagnostic imaging , Gait Disorders, Neurologic/drug therapy , Gait Disorders, Neurologic/etiology , Gait
3.
Mov Disord ; 38(6): 1008-1026, 2023 06.
Article in English | MEDLINE | ID: mdl-37081740

ABSTRACT

BACKGROUND: Antipsychotic-associated movement disorders remain common and disabling. Their screening and assessment are challenging due to clinical heterogeneity and different use of nomenclature between psychiatrists and neurologists. OBJECTIVE: An International Parkinson and Movement Disorder Society subcommittee aimed to rate psychometric quality of severity and screening instruments for antipsychotic-associated movement disorders. METHODS: Following the methodology adopted by previous International Parkinson and Movement Disorders Society subcommittee papers, instruments for antipsychotic-associated movement disorders were reviewed, applying a classification as "recommended," "recommended with caveats," "suggested," or "listed." RESULTS: Our review identified 23 instruments. The highest grade of recommendation reached is "recommended with caveats," assigned to seven severity rating instruments (Extrapyramidal Symptoms Rating Scale, Barnes Akathisia Rating Scale, Abnormal Involuntary Movements Scale, Drug-Induced Extra-Pyramidal Symptoms Scale, Maryland Psychiatric Research Centre involuntary movements scale, Simpson Angus Scale, and Matson Evaluation of Drug Side effects). Only three of these seven (Drug-Induced Extra-Pyramidal Symptoms Scale, Maryland Psychiatric Research Centre, Matson Evaluation of Drug Side effects) were also screening instruments. Their main caveats are insufficient demonstration of psychometric properties (internal consistency, skewing, responsiveness to change) and long duration of administration. Eight "suggested" instruments did not meet requirements for the "recommended" grade also because of insufficient psychometric validation. Other limitations shared by several instruments are lack of comprehensiveness in assessing the spectrum of antipsychotic-associated movement disorders and ambiguous nomenclature. CONCLUSIONS: The high number of instruments "recommended with caveats" does not support the need for developing new instruments for antipsychotic-associated movement disorders. However, addressing the caveats with new psychometric studies and revising existing instruments to improve the clarity of their nomenclature are recommended next steps. © 2023 International Parkinson and Movement Disorder Society.


Subject(s)
Antipsychotic Agents , Basal Ganglia Diseases , Drug-Related Side Effects and Adverse Reactions , Dyskinesia, Drug-Induced , Parkinson Disease , Humans , Antipsychotic Agents/adverse effects , Parkinson Disease/complications , Dyskinesia, Drug-Induced/etiology , Basal Ganglia Diseases/diagnosis
4.
Sensors (Basel) ; 23(19)2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37837160

ABSTRACT

Characterizing motor subtypes of Parkinson's disease (PD) is an important aspect of clinical care that is useful for prognosis and medical management. Although all PD cases involve the loss of dopaminergic neurons in the brain, individual cases may present with different combinations of motor signs, which may indicate differences in underlying pathology and potential response to treatment. However, the conventional method for distinguishing PD motor subtypes involves resource-intensive physical examination by a movement disorders specialist. Moreover, the standardized rating scales for PD rely on subjective observation, which requires specialized training and unavoidable inter-rater variability. In this work, we propose a system that uses machine learning models to automatically and objectively identify some PD motor subtypes, specifically Tremor-Dominant (TD) and Postural Instability and Gait Difficulty (PIGD), from 3D kinematic data recorded during walking tasks for patients with PD (MDS-UPDRS-III Score, 34.7 ± 10.5, average disease duration 7.5 ± 4.5 years). This study demonstrates a machine learning model utilizing kinematic data that identifies PD motor subtypes with a 79.6% F1 score (N = 55 patients with parkinsonism). This significantly outperformed a comparison model using classification based on gait features (19.8% F1 score). Variants of our model trained to individual patients achieved a 95.4% F1 score. This analysis revealed that both temporal, spectral, and statistical features from lower body movements are helpful in distinguishing motor subtypes. Automatically assessing PD motor subtypes simply from walking may reduce the time and resources required from specialists, thereby improving patient care for PD treatments. Furthermore, this system can provide objective assessments to track the changes in PD motor subtypes over time to implement and modify appropriate treatment plans for individual patients as needed.


Subject(s)
Gait Disorders, Neurologic , Parkinson Disease , Humans , Parkinson Disease/pathology , Tremor/diagnosis , Biomechanical Phenomena , Gait , Brain/pathology , Gait Disorders, Neurologic/diagnosis , Postural Balance/physiology
5.
Sensors (Basel) ; 23(4)2023 Feb 04.
Article in English | MEDLINE | ID: mdl-36850363

ABSTRACT

Freezing of gait (FOG) is a poorly understood heterogeneous gait disorder seen in patients with parkinsonism which contributes to significant morbidity and social isolation. FOG is currently measured with scales that are typically performed by movement disorders specialists (i.e., MDS-UPDRS), or through patient completed questionnaires (N-FOG-Q) both of which are inadequate in addressing the heterogeneous nature of the disorder and are unsuitable for use in clinical trials The purpose of this study was to devise a method to measure FOG objectively, hence improving our ability to identify it and accurately evaluate new therapies. A major innovation of our study is that it is the first study of its kind that uses the largest sample size (>30 h, N = 57) in order to apply explainable, multi-task deep learning models for quantifying FOG over the course of the medication cycle and at varying levels of parkinsonism severity. We trained interpretable deep learning models with multi-task learning to simultaneously score FOG (cross-validated F1 score 97.6%), identify medication state (OFF vs. ON levodopa; cross-validated F1 score 96.8%), and measure total PD severity (MDS-UPDRS-III score prediction error ≤ 2.7 points) using kinematic data of a well-characterized sample of N = 57 patients during levodopa challenge tests. The proposed model was able to explain how kinematic movements are associated with each FOG severity level that were highly consistent with the features, in which movement disorders specialists are trained to identify as characteristics of freezing. Overall, we demonstrate that deep learning models' capability to capture complex movement patterns in kinematic data can automatically and objectively score FOG with high accuracy. These models have the potential to discover novel kinematic biomarkers for FOG that can be used for hypothesis generation and potentially as clinical trial outcome measures.


Subject(s)
Gait Disorders, Neurologic , Parkinson Disease , Humans , Gait Disorders, Neurologic/diagnosis , Levodopa/therapeutic use , Parkinson Disease/diagnosis , Gait , Movement
6.
Mov Disord ; 37(2): 253-263, 2022 02.
Article in English | MEDLINE | ID: mdl-34939221

ABSTRACT

Gait and balance abnormalities develop commonly in Parkinson's disease and are among the motor symptoms most disabling and refractory to dopaminergic or other treatments, including deep brain stimulation. Efforts to develop effective therapies are challenged by limited understanding of these complex disorders. There is a major need for novel and appropriately targeted research to expedite progress in this area. The Scientific Issues Committee of the International Parkinson and Movement Disorder Society has charged a panel of experts in the field to consider the current knowledge gaps and determine the research routes with highest potential to generate groundbreaking data. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Gait Disorders, Neurologic , Parkinson Disease , Dopamine , Gait/physiology , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/therapy , Humans , Parkinson Disease/complications , Parkinson Disease/therapy , Research
7.
CNS Spectr ; 27(2): 208-217, 2022 04.
Article in English | MEDLINE | ID: mdl-33213556

ABSTRACT

Accurate diagnosis and appropriate treatment of tardive dyskinesia (TD) are imperative, as its symptoms can be highly disruptive to both patients and their caregivers. Misdiagnosis can lead to incorrect interventions with suboptimal or even deleterious results. To aid in the identification and differentiation of TD in the psychiatric practice setting, we review its clinical features and movement phenomenology, as well as those of other antipsychotic-induced movement disorders, with accompanying links to illustrative videos. Exposure to dopamine receptor blocking agents (DRBAs) such as antipsychotics or antiemetics is associated with a spectrum of movement disorders including TD. The differential diagnosis of TD is based on history of DRBA exposure, recent discontinuation or dose reduction of a DRBA, and movement phenomenology. Common diagnostic challenges are the abnormal behaviors and dyskinesias associated with advanced age or chronic mental illness, and other movement disorders associated with DRBA therapy, such as akathisia, parkinsonian tremor, and tremor related to use of mood stabilizing agents (eg, lithium, divalproex). Duration of exposure may help rule out acute drug-induced syndromes such as acute dystonia or acute/subacute akathisia. Another important consideration is the potential for TD to present together with other drug-induced movement disorders (eg, parkinsonism, parkinsonian tremor, and postural tremor from mood stabilizers) in the same patient, which can complicate both diagnosis and management. After documentation of the phenomenology, severity, and distribution of TD movements, treatment options should be reviewed with the patient and caregivers.


Subject(s)
Antipsychotic Agents , Movement Disorders , Tardive Dyskinesia , Antipsychotic Agents/adverse effects , Humans , Movement Disorders/diagnosis , Movement Disorders/drug therapy , Movement Disorders/etiology , Psychomotor Agitation/drug therapy , Tardive Dyskinesia/chemically induced , Tardive Dyskinesia/diagnosis , Tardive Dyskinesia/drug therapy , Tremor/drug therapy
8.
Mov Disord ; 36(12): 2795-2801, 2021 12.
Article in English | MEDLINE | ID: mdl-34320236

ABSTRACT

BACKGROUND: Several monogenic causes for isolated dystonia have been identified, but they collectively account for only a small proportion of cases. Two genome-wide association studies have reported a few potential dystonia risk loci; but conclusions have been limited by small sample sizes, partial coverage of genetic variants, or poor reproducibility. OBJECTIVE: To identify robust genetic variants and loci in a large multicenter cervical dystonia cohort using a genome-wide approach. METHODS: We performed a genome-wide association study using cervical dystonia samples from the Dystonia Coalition. Logistic and linear regressions, including age, sex, and population structure as covariates, were employed to assess variant- and gene-based genetic associations with disease status and age at onset. We also performed a replication study for an identified genome-wide significant signal. RESULTS: After quality control, 919 cervical dystonia patients compared with 1491 controls of European ancestry were included in the analyses. We identified one genome-wide significant variant (rs2219975, chromosome 3, upstream of COL8A1, P-value 3.04 × 10-8 ). The association was not replicated in a newly genotyped sample of 473 cervical dystonia cases and 481 controls. Gene-based analysis identified DENND1A to be significantly associated with cervical dystonia (P-value 1.23 × 10-6 ). One low-frequency variant was associated with lower age-at-onset (16.4 ± 2.9 years, P-value = 3.07 × 10-8 , minor allele frequency = 0.01), located within the GABBR2 gene on chromosome 9 (rs147331823). CONCLUSION: The genetic underpinnings of cervical dystonia are complex and likely consist of multiple distinct variants of small effect sizes. Larger sample sizes may be needed to provide sufficient statistical power to address the presumably multi-genic etiology of cervical dystonia. © 2021 International Parkinson and Movement Disorder Society.


Subject(s)
Genome-Wide Association Study , Torticollis , Death Domain Receptor Signaling Adaptor Proteins/genetics , Gene Frequency , Genetic Predisposition to Disease/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results , Torticollis/genetics
9.
Brain ; 143(1): 14-30, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31647540

ABSTRACT

Diverse but complementary methodologies are required to uncover the complex determinants and pathophysiology of freezing of gait. To develop future therapeutic avenues, we need a deeper understanding of the disseminated functional-anatomic network and its temporally associated dynamic processes. In this targeted review, we will summarize the latest advances across multiple methodological domains including clinical phenomenology, neurogenetics, multimodal neuroimaging, neurophysiology, and neuromodulation. We found that (i) locomotor network vulnerability is established by structural damage, e.g. from neurodegeneration possibly as result from genetic variability, or to variable degree from brain lesions. This leads to an enhanced network susceptibility, where (ii) modulators can both increase or decrease the threshold to express freezing of gait. Consequent to a threshold decrease, (iii) neuronal integration failure of a multilevel brain network will occur and affect one or numerous nodes and projections of the multilevel network. Finally, (iv) an ultimate pathway might encounter failure of effective motor output and give rise to freezing of gait as clinical endpoint. In conclusion, we derive key questions from this review that challenge this pathophysiological view. We suggest that future research on these questions should lead to improved pathophysiological insight and enhanced therapeutic strategies.


Subject(s)
Brain/physiopathology , Gait Disorders, Neurologic/physiopathology , Parkinson Disease/physiopathology , Apolipoprotein E4/genetics , Brain/diagnostic imaging , Brain Mapping , Cytochrome P-450 CYP2D6/genetics , Functional Neuroimaging , Gait Disorders, Neurologic/diagnostic imaging , Gait Disorders, Neurologic/genetics , Glucosylceramidase/genetics , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Magnetic Resonance Imaging , Mutation , Neural Pathways/physiopathology , Parkinson Disease/diagnostic imaging , Parkinson Disease/genetics , Positron-Emission Tomography , Receptors, Dopamine D2/genetics , Tomography, Emission-Computed, Single-Photon
10.
J Neurol Neurosurg Psychiatry ; 91(11): 1210-1218, 2020 11.
Article in English | MEDLINE | ID: mdl-32732384

ABSTRACT

Loss of nigrostriatal dopaminergic projection neurons is a key pathology in Parkinson's disease, leading to abnormal function of basal ganglia motor circuits and the accompanying characteristic motor features. A number of intraparenchymally delivered gene therapies designed to modify underlying disease and/or improve clinical symptoms have shown promise in preclinical studies and subsequently were evaluated in clinical trials. Here we review the challenges with surgical delivery of gene therapy vectors that limited therapeutic outcomes in these trials, particularly the lack of real-time monitoring of vector administration. These challenges have recently been addressed during the evolution of novel techniques for vector delivery that include the use of intraoperative MRI. The preclinical development of these techniques are described in relation to recent clinical translation in an adeno-associated virus serotype 2-mediated human aromatic L-amino acid decarboxylase gene therapy development programme. This new paradigm allows visualisation of the accuracy and adequacy of viral vector delivery within target structures, enabling intertrial modifications in surgical approaches, cannula design, vector volumes and dosing. The rapid, data-driven evolution of these procedures is unique and has led to improved vector delivery.


Subject(s)
Corpus Striatum , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Magnetic Resonance Imaging , Neurosurgical Procedures/methods , Parkinson Disease/therapy , Substantia Nigra , Animals , Aromatic-L-Amino-Acid Decarboxylases/genetics , Basal Ganglia , Dependovirus , Evidence-Based Medicine , GTP Cyclohydrolase/genetics , Glutamate Decarboxylase/genetics , Humans , Intraoperative Care/methods , Lentivirus , Neurturin/genetics , Parvovirinae , Primates , Surgery, Computer-Assisted , Tyrosine 3-Monooxygenase/genetics
11.
CNS Spectr ; : 1-8, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33161935

ABSTRACT

Functional neurological disorder (FND) is a complex neuropsychiatric syndrome with many phenotypes that are commonly encountered in clinical practice. Despite the heterogeneity of FND, the rate of misidentification is consistently low. For the more common motor subtypes, there are clear positive clinical, electrophysiological, and rarely imaging criteria that can establish the diagnosis in the traditional sense. For nonmotor subtypes, the characterization may be less clear. Here, we argue that the current diagnostic criteria are not reflective of the current shared neuropsychiatric understanding of FND, and, as a result, provide an incomplete picture of the diagnosis. We propose a three-step diagnostic triad for FND, in which the traditional neurological diagnosis is only the first element. Other steps include psychiatric/psychological formulation, integration, and follow-up. We advocate that this diagnostic approach should be the shared responsibility of neurology and mental health professionals. Finally, a research agenda is proposed to address the missing factors in the field.

12.
J Neurol Neurosurg Psychiatry ; 90(12): 1317-1323, 2019 12.
Article in English | MEDLINE | ID: mdl-31296586

ABSTRACT

OBJECTIVE: To evaluate the long-term safety and efficacy of deutetrabenazine in patients with tardive dyskinesia (TD). METHOD: Patients with TD who completed the 12 week, phase 3, placebo-controlled trials were eligible to enter this open-label, single-arm study. The open-label study consisted of a 6 week dose-escalation phase and a long-term maintenance phase (clinic visits at Weeks 4, 6 and 15, and every 13 weeks until Week 106). Patients began deutetrabenazine at 12 mg/day, titrating up to a dose that was tolerable and provided adequate dyskinesia control, based on investigator judgement, with a maximum allowed dose of 48 mg/day (36 mg/day for patients taking strong cytochrome P450 2D6 (CYP2D6) inhibitors). Safety measures included incidence of adverse events (AEs) and scales used to monitor parkinsonism, akathisia/restlessness, anxiety, depression, suicidality and somnolence/sedation. Efficacy endpoints included the change in Abnormal Involuntary Movement Scale (AIMS) score (items 1 to 7) from baseline and the proportion of patients rated as 'Much Improved' or 'Very Much Improved' on the Clinical Global Impression of Change. RESULTS: A total of 343 patients enrolled in the extension study, and there were 331 patient-years of exposure in this analysis. The exposure-adjusted incidence rates of AEs with long-term treatment were comparable to or lower than those observed in the phase 3 trials. The mean (SE) change in AIMS score was -4.9 (0.4) at Week 54 (n = 146), - 6.3 (0.7) at Week 80 (n = 66) and -5.1 (2.0) at Week 106 (n = 8). CONCLUSIONS: Overall, long-term treatment with deutetrabenazine was efficacious, safe, and well tolerated in patients with TD. TRIAL REGISTRATION NUMBER: NCT02198794.


Subject(s)
Anti-Dyskinesia Agents/therapeutic use , Tardive Dyskinesia/drug therapy , Tetrabenazine/analogs & derivatives , Adult , Aged , Anti-Dyskinesia Agents/adverse effects , Antipsychotic Agents/adverse effects , Cytochrome P-450 CYP2D6 Inhibitors/adverse effects , Cytochrome P-450 CYP2D6 Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Mood Disorders/complications , Mood Disorders/drug therapy , Psychotic Disorders/complications , Psychotic Disorders/drug therapy , Tardive Dyskinesia/physiopathology , Tetrabenazine/adverse effects , Tetrabenazine/therapeutic use , Treatment Outcome
13.
Mov Disord ; 34(3): 416-419, 2019 03.
Article in English | MEDLINE | ID: mdl-30597635

ABSTRACT

BACKGROUND: Previous studies investigating nigral iron accumulation used T2 or T2 *-weighted contrasts to define the regions of interest (ROIs) in the substantia nigra with mixed results. Because these contrasts are not sensitive to neuromelanin, ROIs may have inadvertently missed the SNpc. An approach sensitive to neuromelanin should yield consistent results. We examine iron deposition in ROIs derived from neuromelanin-sensitive and T2 *-weighted contrasts, respectively. METHODS: T1 -weighted and multiecho gradient echo imaging data were obtained in 2 cohorts. Multiecho gradient echo imaging data were analyzed using neuromelanin-sensitive SNpc ROIs as well as T2 *-weighted SNr ROIs. RESULTS: When compared with controls, significantly larger R2 * values were seen in the SNpc of PD patients in both cohorts. Mean R2 * values in the SNr of PD patients showed no consistency, with 1 cohort showing a small, statistically significant increase, whereas the other cohort exhibited no statistical difference. CONCLUSION: Mean R2 * in the SNpc defined by neuromelanin-sensitive MRI is significantly increased in PD. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Iron/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Aged , Female , Humans , Magnetic Resonance Imaging , Male , Melanins , Middle Aged , Parkinson Disease/diagnostic imaging , Parkinson Disease/pathology , Substantia Nigra/diagnostic imaging , Substantia Nigra/pathology
14.
Hum Mol Genet ; 25(17): 3849-3862, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27402877

ABSTRACT

Parkinson's disease (PD) is the most common cause of neurodegenerative movement disorder and the second most common cause of dementia. Genes are thought to have a stronger effect on age-at-onset of PD than on risk, yet there has been a phenomenal success in identifying risk loci but not age-at-onset modifiers. We conducted a genome-wide study for age-at-onset. We analysed familial and non-familial PD separately, per prior evidence for strong genetic effect on age-at-onset in familial PD. GWAS was conducted in 431 unrelated PD individuals with at least one affected relative (familial PD) and 1544 non-familial PD from the NeuroGenetics Research Consortium (NGRC); an additional 737 familial PD and 2363 non-familial PD were used for replication. In familial PD, two signals were detected and replicated robustly: one mapped to LHFPL2 on 5q14.1 (PNGRC = 3E-8, PReplication = 2E-5, PNGRC + Replication = 1E-11), the second mapped to TPM1 on 15q22.2 (PNGRC = 8E-9, PReplication = 2E-4, PNGRC + Replication = 9E-11). The variants that were associated with accelerated onset had low frequencies (<0.02). The LHFPL2 variant was associated with earlier onset by 12.33 [95% CI: 6.2; 18.45] years in NGRC, 8.03 [2.95; 13.11] years in replication, and 9.79 [5.88; 13.70] years in the combined data. The TPM1 variant was associated with earlier onset by 15.30 [8.10; 22.49] years in NGRC, 9.29 [1.79; 16.79] years in replication, and 12.42 [7.23; 17.61] years in the combined data. Neither LHFPL2 nor TPM1 was associated with age-at-onset in non-familial PD. LHFPL2 (function unknown) is overexpressed in brain tumours. TPM1 encodes a highly conserved protein that regulates muscle contraction, and is a tumour-suppressor gene.


Subject(s)
Genome-Wide Association Study/methods , Membrane Proteins/genetics , Parkinson Disease/genetics , Proteins/genetics , Tropomyosin/genetics , Age of Onset , Aged , Aged, 80 and over , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
15.
Mov Disord ; 33(5): 793-804, 2018 05.
Article in English | MEDLINE | ID: mdl-29572994

ABSTRACT

BACKGROUND: Gastrointestinal symptoms are common in Parkinson's disease and frequently precede the development of motor impairments. Intestinal inflammation has been proposed as a driver of disease pathology, and evaluation of inflammatory mediators in stool could possibly identify valuable early-stage biomarkers. We measured immune- and angiogenesis-related proteins in human stool to examine inflammatory profiles associated with Parkinson's disease. METHODS: Stool samples and subjects' self-reported metadata were obtained from 156 individuals with Parkinson's disease and 110 without, including spouse and nonhousehold controls. Metadata were probed for disease-associated differences, and levels of 37 immune and angiogenesis factors in stool homogenates were measured by multiplexed immunoassay and compared across experimental groups. RESULTS: Parkinson's disease patients reported greater incidence of intestinal disease and digestive problems than controls. Direct comparison of levels of stool analytes in patients and controls revealed elevated vascular endothelial growth factor receptor 1, interleukin-1α, and CXCL8 in patients' stool. Paired comparison of patients and spouses suggested higher levels of multiple factors in patients, but this was complicated by sex differences. Sex, body mass index, a history of smoking, and use of probiotics were found to strongly influence levels of stool analytes. Multivariate analysis accounting for these and other potential confounders confirmed elevated levels of interleukin-1α and CXCL8 and also revealed increased interleukin-1ß and C-reactive protein in stool in Parkinson's disease. These differences were not dependent on subject age or disease duration. CONCLUSIONS: Levels of stool immune factors indicate that intestinal inflammation is present in patients with Parkinson's disease. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Cytokines/metabolism , Feces/chemistry , Gastroenteritis/etiology , Gastroenteritis/metabolism , Parkinson Disease/complications , Adult , Aged , Aged, 80 and over , Angiogenesis Inducing Agents/metabolism , C-Reactive Protein/metabolism , Female , Humans , Male , Middle Aged , Mood Disorders/etiology , Parkinson Disease/psychology , Sex Characteristics
16.
Mov Disord ; 33(11): 1769-1776, 2018 11.
Article in English | MEDLINE | ID: mdl-30387904

ABSTRACT

BACKGROUND: PD psychosis is often associated with cognitive impairment, including dementia, and involves dopaminergic, serotonergic, and cholinergic mechanisms. OBJECTIVE: To evaluate the differential effect of the antipsychotic pimavanserin, a selective serotonin 2A receptor inverse agonist, in PD psychosis patients with versus without cognitive impairment and in those receiving versus not receiving cognitive-enhancing medications. METHODS: Data from the pivotal randomized clinical trial of pimavanserin for PD psychosis were stratified by (1) screening MMSE score as cognitively impaired (21-24) versus unimpaired (≥25) and (2) concomitant use versus nonuse of cognitive-enhancing medications. The primary outcome measure was change in the PD-adapted Scale for the Assessment of Positive Symptoms. RESULTS: Mean (pimavanserin vs. placebo) change from baseline was larger in the cognitively impaired (n = 50; -6.62 vs. -0.91; P = 0.002) versus the cognitively unimpaired (n = 135; -5.50 vs. -3.23; p = 0.046) group. The comparable change was -6.04 versus -2.18 (P = 0.012) and -5.66 versus -3.15 (P = 0.041) in patients treated (n = 69) and not treated (n = 116) with concomitant cognitive-enhancing medication. Pimavanserin was similarly tolerated across all cognitive groups with no additional safety concerns identified. Overall adverse event rates were comparable across the concomitant cognitive-enhancing medication groups; however, rates of serious adverse events and discontinuations attributed to adverse events were increased in patients taking cholinesterase inhibitors. CONCLUSIONS: The antipsychotic effect of pimavanserin is robust in PD patients with cognitive impairment and may be enhanced by concomitant cognitive-enhancing medication use. Future prospective studies are needed to confirm these preliminary findings. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Antipsychotic Agents/therapeutic use , Cognition Disorders/drug therapy , Nootropic Agents/therapeutic use , Parkinson Disease/complications , Piperidines/therapeutic use , Psychotic Disorders/drug therapy , Psychotic Disorders/etiology , Urea/analogs & derivatives , Aged , Aged, 80 and over , Cognition Disorders/etiology , Double-Blind Method , Female , Humans , Male , Mental Status Schedule , Middle Aged , Treatment Outcome , Urea/therapeutic use
17.
Hum Brain Mapp ; 38(5): 2627-2634, 2017 05.
Article in English | MEDLINE | ID: mdl-28240402

ABSTRACT

The objective of this study was to measure neuromelanin-sensitive MRI contrast changes in the lateral-ventral tier of substantia nigra pars compacta in Parkinson's disease (PD). Histopathological studies of PD have demonstrated both massive loss of melanized dopamine neurons and iron accumulation in the substantia nigra pars compacta. Neurodegeneration is most profound in the lateral-ventral tier of this structure. We have previously shown in both healthy controls and individuals with PD that neuromelanin-sensitive MRI and iron-sensitive MRI contrast regions in substantia nigra overlap. This overlap region is located in the lateral-ventral tier. Exploiting this area of contrast overlap for region of interest selection, we developed a semi-automated image processing approach to characterize the lateral-ventral tier in MRI data. Here we apply this approach to measure magnetization transfer contrast, which corresponds to local neuromelanin density, in both the lateral-ventral tier and the entire pars compacta in 22 PD patients and 19 controls. Significant contrast reductions were seen in PD in both the entire pars compacta (P = 0.009) and in its lateral-ventral tier (P = 0.0002); in PD contrast was significantly lower in the lateral-ventral tier than in the entire pars compacta (P = 0.0008). These findings are the first in vivo evidence of the selective vulnerability of this nigral subregion in PD, and this approach may be developed for high impact biomarker applications. Hum Brain Mapp 38:2627-2634, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/pathology , Parkinson Disease/complications , Substantia Nigra/pathology , Aged , Analysis of Variance , Cohort Studies , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , ROC Curve , Severity of Illness Index , Substantia Nigra/diagnostic imaging , Surveys and Questionnaires
18.
Ann Neurol ; 80(5): 644-659, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27649270

ABSTRACT

Freezing of gait (FOG) is a common and debilitating, but largely mysterious, symptom of Parkinson disease. In this review, we will discuss the cerebral substrate of FOG focusing on brain physiology and animal models. Walking is a combination of automatic movement processes, afferent information processing, and intentional adjustments. Thus, normal gait requires a delicate balance between various interacting neuronal systems. To further understand gait control and specifically FOG, we will discuss the basic physiology of gait, animal models of gait disturbance including FOG, alternative etiologies of FOG, and functional magnetic resonance studies investigating FOG. The outcomes of these studies point to a dynamic network of cortical areas such as the supplementary motor area, as well as subcortical areas such as the striatum and the mesencephalic locomotor region including the pedunculopontine nucleus (PPN). Additionally, we will review PPN (area) stimulation as a possible treatment for FOG, and ponder whether PPN stimulation truly is the right step forward. Ann Neurol 2016;80:644-659.


Subject(s)
Deep Brain Stimulation , Functional Neuroimaging , Gait Disorders, Neurologic , Pedunculopontine Tegmental Nucleus , Animals , Gait Disorders, Neurologic/etiology , Gait Disorders, Neurologic/physiopathology , Gait Disorders, Neurologic/therapy , Humans
20.
Mov Disord ; 32(6): 893-903, 2017 06.
Article in English | MEDLINE | ID: mdl-28370447

ABSTRACT

BACKGROUND: Nondopaminergic pathways represent potential targets to treat levodopa-induced dyskinesia in Parkinson's disease (PD). This pilot-study (NCT01767129) examined the safety/efficacy of the sigma-1 receptor-agonist and glutamatergic/monoaminergic modulator, dextromethorphan plus quinidine (to inhibit rapid dextromethorphan metabolism), for treating levodopa-induced dyskinesia. METHODS: PD patients were randomized to dextromethorphan/quinidine (45 mg/10 mg twice daily)/placebo in two 2-week double-blind, crossover treatment periods, with intervening 2-week washout. After 14 days, a 2-hour intravenous levodopa-infusion was administered. Patient examinations were videotaped before infusion ("off" state) and every 30 minutes during and afterwards until patients returned to "off." The primary endpoint was dyskinesia-severity during infusion measured by Unified Dyskinesia Rating Scale part 3 area-under-curve scores (blinded expert rated). Additional endpoints included other dyskinesia/motor assessments, global measures of clinical-change, and adverse-events. RESULTS: A total of 13 patients were randomized and completed the study (efficacy-evaluable population). Dyskinesia-severity was nonsignificantly lower with dextromethorphan/quinidine than placebo during infusion (area-under-curve 966.5 vs 1048.8; P = .191 [efficacy-evaluable patients]), and significantly lower in a post-hoc sensitivity analysis of the per-protocol-population (efficacy-evaluable patients with ≥ 80% study-drug-compliance, n = 12) when measured from infusion start to 4-hours post-infusion completion (area-under-curve 1585.0 vs 1911.3; P = .024). Mean peak dyskinesia decreased significantly from infusion-start to return to "off" (13.3 vs 14.9; P = .018 [efficacy-evaluable patients]). A total of 9 patients rated dyskinesia "much/very much improved" on dextromethorphan/quinidine versus 1-patient on placebo. Dextromethorphan/quinidine did not worsen PD-motor scores, was generally well tolerated, and was associated with more frequent adverse events. CONCLUSION: This study provides preliminary evidence of clinical benefit with dextromethorphan/quinidine for treating levodopa-induced dyskinesia in PD. Larger studies with a longer treatment duration need to corroborate these early findings. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/adverse effects , Dextromethorphan/pharmacology , Dyskinesia, Drug-Induced/drug therapy , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Levodopa/adverse effects , Outcome Assessment, Health Care , Parkinson Disease/drug therapy , Quinidine/pharmacology , Aged , Cross-Over Studies , Dextromethorphan/administration & dosage , Dextromethorphan/adverse effects , Double-Blind Method , Drug Therapy, Combination , Dyskinesia, Drug-Induced/etiology , Enzyme Inhibitors/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/adverse effects , Female , Humans , Male , Middle Aged , Pilot Projects , Quinidine/administration & dosage , Quinidine/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL