1
|
Bentall L, Parr‐Brownlie L. Sexual Dimorphism in Levodopa-Induced Dyskinesia Following Parkinson's Disease: Uncharted Territory. Eur J Neurosci 2025; 61:e70144. [PMID: 40360439 PMCID: PMC12075048 DOI: 10.1111/ejn.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Sexual dimorphism is well-documented in Parkinson's disease (PD); however, when it comes to levodopa-induced dyskinesia (LID), epidemiological and clinical findings are scarce. This is an oversight because recent studies show significant correlations between LID risk and female sex. Estrogen strongly impacts neuronal function, affecting cognitive tasks such as movement, object recognition, and reward. In movement pathways, estrogen increases dopamine synthesis, transmission, and regulation, resulting in neuroprotection for PD in women. However, following menopause, PD prevalence, symptom severity, and LID risk increase for women. Consequently, early to mid-life estrogen state is neuroprotective, but later in life becomes a risk factor for PD and LID. This review explores estrogen's action in the brain, specifically within the dopamine system. Sexual dimorphism is described for the prevalence and onset of PD and LID. We examine the cellular basis of estrogen's role in sexual dimorphism and integrate these ideas to hypothesize why the risk for LID is higher for women, than men, with PD. Lastly, this review proposes that women with PD need their symptoms to be considered and managed differently to males. Treatment of women with PD should be based on their menopausal stage, as estrogen may be masking, exacerbating, or complicating symptoms. Importantly, we present these concepts to stimulate discussion among clinical and bench scientists so that key experiments can be conducted to examine the mechanisms underlying LID, so they can be prevented to improve the quality of life for women and men living with PD in the future.
Collapse
|
2
|
Nakano Y, Hirano S, Tamura M, Koizumi Y, Kitayama Y, Izumi M, Suzuki M, Yamagishi K, Suzuki Y, Ikeda S, Arase A, Yamamoto T, Yamanaka Y, Sugiyama A, Shibuya K, Horikoshi T, Uno T, Eidelberg D, Kuwabara S. Clinical and brain functional correlates of instrumental rigidity measurement in Parkinson's disease. Sci Rep 2025; 15:8056. [PMID: 40055432 PMCID: PMC11889231 DOI: 10.1038/s41598-025-91567-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/21/2025] [Indexed: 03/12/2025] Open
Abstract
Rigidity, a cardinal symptom of Parkinson's disease (PD), remains challenging to assess objectively. A torque-angle instrument was developed to quantify muscle tone, providing two parameters: bias difference and elastic coefficient. This study aimed to investigate the association of the instrument-measured rigidity with clinical assessments and brain function. In 30 patients with PD, the muscle tone in both arms was evaluated. Ten with wearing-off phenomenon were assessed twice, off and on condition. Twentynine patients underwent brain perfusion single-photon emission computed tomography (SPECT), and expression of PD-related covariance pattern (PDRP) was computed. Bias difference and elastic coefficient showed positive correlations with physician-rated rigidity (P < 0.002). Bias difference decreased after dopaminergic medication (P = 0.022) and was associated with lower body mass index (P = 0.012). Elastic coefficient positively correlated with the Unified PD Rating Scale Part III and PDRP scores (P < 0.044). Furthermore, the higher bias difference correlated with decreased sensory-motor cortex and increased substantia nigra perfusion (P < 0.001). The Torque-angle instrument is a viable tool for quantifying rigidity in PD. The bias difference reflects treatment responsiveness and is associated with the function in the sensory-motor cortex and substantia nigra. The elastic coefficient is indicative of overall Parkinsonism severity.
Collapse
Affiliation(s)
- Yoshikazu Nakano
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Shigeki Hirano
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan.
| | - Mitsuyoshi Tamura
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Yume Koizumi
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Yoshihisa Kitayama
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Michiko Izumi
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Masahide Suzuki
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Kosuke Yamagishi
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Yutaro Suzuki
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Shinobu Ikeda
- Division of Laboratory Medicine, Chiba University Hospital, Chiba, Chiba, Japan
| | - Ayaka Arase
- Division of Laboratory Medicine, Chiba University Hospital, Chiba, Chiba, Japan
| | - Tatsuya Yamamoto
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
- Division of Occupational Therapy, Department of Rehabilitation, Chiba Prefectural University of Health Sciences, Chiba, Chiba, Japan
| | - Yoshitaka Yamanaka
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
- Urayasu Rehabilitation Education Center, Chiba University Hospital, Urayasu, Chiba, Japan
- Department of Rehabilitation, Kimitsu Chuo Hospital, Kimitsu, Chiba, Japan
| | - Atsuhiko Sugiyama
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Kazumoto Shibuya
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| | - Takuro Horikoshi
- Diagnostic Radiology and Radiation Oncology, Chiba University Graduate School of Medicine, Chiba, Chiba, Japan
| | - Takashi Uno
- Diagnostic Radiology and Radiation Oncology, Chiba University Graduate School of Medicine, Chiba, Chiba, Japan
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Satoshi Kuwabara
- Department of Neurology, Chiba University Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba, 264-8670, Chiba, Japan
| |
Collapse
|
3
|
Unadkat P, Vo A, Ma Y, Peng S, Nguyen N, Niethammer M, Tang CC, Dhawan V, Ramdhani R, Fenoy A, Caminiti SP, Perani D, Eidelberg D. Deep brain stimulation of the subthalamic nucleus for Parkinson's disease: A network imaging marker of the treatment response. RESEARCH SQUARE 2024:rs.3.rs-4178280. [PMID: 38766007 PMCID: PMC11100869 DOI: 10.21203/rs.3.rs-4178280/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Subthalamic nucleus deep brain stimulation (STN-DBS) alleviates motor symptoms of Parkinson's disease (PD), thereby improving quality of life. However, quantitative brain markers to evaluate DBS responses and select suitable patients for surgery are lacking. Here, we used metabolic brain imaging to identify a reproducible STN-DBS network for which individual expression levels increased with stimulation in proportion to motor benefit. Of note, measurements of network expression from metabolic and BOLD imaging obtained preoperatively predicted motor outcomes determined after DBS surgery. Based on these findings, we computed network expression in 175 PD patients, with time from diagnosis ranging from 0 to 21 years, and used the resulting data to predict the outcome of a potential STN-DBS procedure. While minimal benefit was predicted for patients with early disease, the proportion of potential responders increased after 4 years. Clinically meaningful improvement with stimulation was predicted in 18.9 - 27.3% of patients depending on disease duration.
Collapse
Affiliation(s)
| | - An Vo
- The Feinstein Institutes for Medical Research
| | - Yilong Ma
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Shichun Peng
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | | | | | | | | | - Ritesh Ramdhani
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell
| | | | | | | | | |
Collapse
|
4
|
Wu C, Wu H, Zhou C, Guan X, Guo T, Wu J, Chen J, Wen J, Qin J, Tan S, Duanmu X, Yuan W, Zheng Q, Zhang B, Xu X, Zhang M. Neurovascular coupling alteration in drug-naïve Parkinson's disease: The underlying molecular mechanisms and levodopa's restoration effects. Neurobiol Dis 2024; 191:106406. [PMID: 38199273 DOI: 10.1016/j.nbd.2024.106406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) patients exhibit an imbalance between neuronal activity and perfusion, referred to as abnormal neurovascular coupling (NVC). Nevertheless, the underlying molecular mechanism and how levodopa, the standard treatment in PD, regulates NVC is largely unknown. MATERIAL AND METHODS A total of 52 drug-naïve PD patients and 49 normal controls (NCs) were enrolled. NVC was characterized in vivo by relating cerebral blood flow (CBF) and amplitude of low-frequency fluctuations (ALFF). Motor assessments and MRI scanning were conducted on drug-naïve patients before and after levodopa therapy (OFF/ON state). Regional NVC differences between patients and NCs were identified, followed by an assessment of the associated receptors/transporters. The influence of levodopa on NVC, CBF, and ALFF within these abnormal regions was analyzed. RESULTS Compared to NCs, OFF-state patients showed NVC dysfunction in significantly lower NVC in left precentral, postcentral, superior parietal cortex, and precuneus, along with higher NVC in left anterior cingulate cortex, right olfactory cortex, thalamus, caudate, and putamen (P-value <0.0006). The distribution of NVC differences correlated with the density of dopaminergic, serotonin, MU-opioid, and cholinergic receptors/transporters. Additionally, levodopa ameliorated abnormal NVC in most of these regions, where there were primarily ALFF changes with limited CBF modifications. CONCLUSION Patients exhibited NVC dysfunction primarily in the striato-thalamo-cortical circuit and motor control regions, which could be driven by dopaminergic and nondopaminergic systems, and levodopa therapy mainly restored abnormal NVC by modulating neuronal activity.
Collapse
Affiliation(s)
- Chenqing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoting Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhou
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingwen Chen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmei Qin
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sijia Tan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojie Duanmu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijin Yuan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianshi Zheng
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
5
|
Barbero JA, Unadkat P, Choi YY, Eidelberg D. Functional Brain Networks to Evaluate Treatment Responses in Parkinson's Disease. Neurotherapeutics 2023; 20:1653-1668. [PMID: 37684533 PMCID: PMC10684458 DOI: 10.1007/s13311-023-01433-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Network analysis of functional brain scans acquired with [18F]-fluorodeoxyglucose positron emission tomography (FDG PET, to map cerebral glucose metabolism), or resting-state functional magnetic resonance imaging (rs-fMRI, to map blood oxygen level-dependent brain activity) has increasingly been used to identify and validate reproducible circuit abnormalities associated with neurodegenerative disorders such as Parkinson's disease (PD). In addition to serving as imaging markers of the underlying disease process, these networks can be used singly or in combination as an adjunct to clinical diagnosis and as a screening tool for therapeutics trials. Disease networks can also be used to measure rates of progression in natural history studies and to assess treatment responses in individual subjects. Recent imaging studies in PD subjects scanned before and after treatment have revealed therapeutic effects beyond the modulation of established disease networks. Rather, other mechanisms of action may be at play, such as the induction of novel functional brain networks directly by treatment. To date, specific treatment-induced networks have been described in association with novel interventions for PD such as subthalamic adeno-associated virus glutamic acid decarboxylase (AAV2-GAD) gene therapy, as well as sham surgery or oral placebo under blinded conditions. Indeed, changes in the expression of these networks with treatment have been found to correlate consistently with clinical outcome. In aggregate, these attributes suggest a role for functional brain networks as biomarkers in future clinical trials.
Collapse
Affiliation(s)
- János A Barbero
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Prashin Unadkat
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA
| | - Yoon Young Choi
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Molecular Medicine and Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA.
| |
Collapse
|
6
|
Spetsieris PG, Eidelberg D. Parkinson's disease progression: Increasing expression of an invariant common core subnetwork. Neuroimage Clin 2023; 39:103488. [PMID: 37660556 PMCID: PMC10491857 DOI: 10.1016/j.nicl.2023.103488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Notable success has been achieved in the study of neurodegenerative conditions using reduction techniques such as principal component analysis (PCA) and sparse inverse covariance estimation (SICE) in positron emission tomography (PET) data despite their widely differing approach. In a recent study of SICE applied to metabolic scans from Parkinson's disease (PD) patients, we showed that by using PCA to prespecify disease-related partition layers, we were able to optimize maps of functional metabolic connectivity within the relevant networks. Here, we show the potential of SICE, enhanced by disease-specific subnetwork partitions, to identify key regional hubs and their connections, and track their associations in PD patients with increasing disease duration. This approach enabled the identification of a core zone that included elements of the striatum, pons, cerebellar vermis, and parietal cortex and provided a deeper understanding of progressive changes in their connectivity. This subnetwork constituted a robust invariant disease feature that was unrelated to phenotype. Mean expression levels for this subnetwork increased steadily in a group of 70 PD patients spanning a range of symptom durations between 1 and 21 years. The findings were confirmed in a validation sample of 69 patients with up to 32 years of symptoms. The common core elements represent possible targets for disease modification, while their connections to external regions may be better suited for symptomatic treatment.
Collapse
Affiliation(s)
- Phoebe G Spetsieris
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, United States
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, United States; Molecular Medicine and Neurology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, United States.
| |
Collapse
|
7
|
Irmady K, Hale CR, Qadri R, Fak J, Simelane S, Carroll T, Przedborski S, Darnell RB. Blood transcriptomic signatures associated with molecular changes in the brain and clinical outcomes in Parkinson's disease. Nat Commun 2023; 14:3956. [PMID: 37407548 DOI: 10.1038/s41467-023-39652-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023] Open
Abstract
The ability to use blood to predict the outcomes of Parkinson's disease, including disease progression and cognitive and motor complications, would be of significant clinical value. We undertook bulk RNA sequencing from the caudate and putamen of postmortem Parkinson's disease (n = 35) and control (n = 40) striatum, and compared molecular profiles with clinical features and bulk RNA sequencing data obtained from antemortem peripheral blood. Cognitive and motor complications of Parkinson's disease were associated with molecular changes in the caudate (stress response) and putamen (endothelial pathways) respectively. Later and earlier-onset Parkinson's disease were molecularly distinct, and disease duration was associated with changes in caudate (oligodendrocyte development) and putamen (cellular senescence), respectively. Transcriptome patterns in the postmortem Parkinson's disease brain were also evident in antemortem peripheral blood, and correlated with clinical features of the disease. Together, these findings identify molecular signatures in Parkinson's disease patients' brain and blood of potential pathophysiologic and prognostic importance.
Collapse
Affiliation(s)
- Krithi Irmady
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| | - Caryn R Hale
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Rizwana Qadri
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - John Fak
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Sitsandziwe Simelane
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Thomas Carroll
- Bioinformatics Resource Center, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Pathology & Cell Biology, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neuroscience, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Robert B Darnell
- Laboratory of Molecular Neuro-oncology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
- Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
8
|
di Biase L, Pecoraro PM, Carbone SP, Caminiti ML, Di Lazzaro V. Levodopa-Induced Dyskinesias in Parkinson's Disease: An Overview on Pathophysiology, Clinical Manifestations, Therapy Management Strategies and Future Directions. J Clin Med 2023; 12:4427. [PMID: 37445461 DOI: 10.3390/jcm12134427] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/18/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Since its first introduction, levodopa has become the cornerstone for the treatment of Parkinson's disease and remains the leading therapeutic choice for motor control therapy so far. Unfortunately, the subsequent appearance of abnormal involuntary movements, known as dyskinesias, is a frequent drawback. Despite the deep knowledge of this complication, in terms of clinical phenomenology and the temporal relationship during a levodopa regimen, less is clear about the pathophysiological mechanisms underpinning it. As the disease progresses, specific oscillatory activities of both motor cortical and basal ganglia neurons and variation in levodopa metabolism, in terms of the dopamine receptor stimulation pattern and turnover rate, underlie dyskinesia onset. This review aims to provide a global overview on levodopa-induced dyskinesias, focusing on pathophysiology, clinical manifestations, therapy management strategies and future directions.
Collapse
Affiliation(s)
- Lazzaro di Biase
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Brain Innovations Lab, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Pasquale Maria Pecoraro
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Simona Paola Carbone
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Maria Letizia Caminiti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Vincenzo Di Lazzaro
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Unit of Neurology, Neurophysiology, Neurobiology and Psichiatry, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
9
|
Perovnik M, Rus T, Schindlbeck KA, Eidelberg D. Functional brain networks in the evaluation of patients with neurodegenerative disorders. Nat Rev Neurol 2023; 19:73-90. [PMID: 36539533 DOI: 10.1038/s41582-022-00753-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Network analytical tools are increasingly being applied to brain imaging maps of resting metabolic activity (PET) or blood oxygenation-dependent signals (functional MRI) to characterize the abnormal neural circuitry that underlies brain diseases. This approach is particularly valuable for the study of neurodegenerative disorders, which are characterized by stereotyped spread of pathology along discrete neural pathways. Identification and validation of disease-specific brain networks facilitate the quantitative assessment of pathway changes over time and during the course of treatment. Network abnormalities can often be identified before symptom onset and can be used to track disease progression even in the preclinical period. Likewise, network activity can be modulated by treatment and might therefore be used as a marker of efficacy in clinical trials. Finally, early differential diagnosis can be achieved by simultaneously measuring the activity levels of multiple disease networks in an individual patient's scans. Although these techniques were originally developed for PET, over the past several years analogous methods have been introduced for functional MRI, a more accessible non-invasive imaging modality. This advance is expected to broaden the application of network tools to large and diverse patient populations.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tomaž Rus
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
10
|
Kokkonen A, Honkanen EA, Corp DT, Joutsa J. Neurobiological effects of deep brain stimulation: A systematic review of molecular brain imaging studies. Neuroimage 2022; 260:119473. [PMID: 35842094 DOI: 10.1016/j.neuroimage.2022.119473] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/28/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Deep brain stimulation (DBS) is an established treatment for several brain disorders, including Parkinson's disease, essential tremor, dystonia and epilepsy, and an emerging therapeutic tool in many other neurological and psychiatric disorders. The therapeutic efficacy of DBS is dependent on the stimulation target, but its mechanisms of action are still relatively poorly understood. Investigating these mechanisms is challenging, partly because the stimulation devices and electrodes have limited the use of functional MRI in these patients. Molecular brain imaging techniques, such as positron emission tomography (PET) and single photon emission tomography (SPET), offer a unique opportunity to characterize the whole brain effects of DBS. Here, we investigated the direct effects of DBS by systematically reviewing studies performing an `on' vs `off' contrast during PET or SPET imaging. We identified 62 studies (56 PET and 6 SPET studies; 531 subjects). Approximately half of the studies focused on cerebral blood flow or glucose metabolism in patients Parkinson's disease undergoing subthalamic DBS (25 studies, n = 289), therefore Activation Likelihood Estimation analysis was performed on these studies. Across disorders and stimulation targets, DBS was associated with a robust local increase in ligand uptake at the stimulation site and target-specific remote network effects. Subthalamic nucleus stimulation in Parkinson's disease showed a specific pattern of changes in the motor circuit, including increased ligand uptake in the basal ganglia, and decreased ligand uptake in the primary motor cortex, supplementary motor area and cerebellum. However, there was only a handful of studies investigating other brain disorder and stimulation site combinations (1-3 studies each), or specific neurotransmitter systems, preventing definitive conclusions of the detailed molecular effects of the stimulation in these cases.
Collapse
Affiliation(s)
- Aleksi Kokkonen
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Turku, Finland; Turku PET Center, Neurocenter, Turku University Hospital, Turku, Finland.
| | - Emma A Honkanen
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Turku, Finland; Turku PET Center, Neurocenter, Turku University Hospital, Turku, Finland
| | - Daniel T Corp
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Juho Joutsa
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Turku, Finland; Turku PET Center, Neurocenter, Turku University Hospital, Turku, Finland; Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, Boston, MA, United States of America.
| |
Collapse
|
11
|
Nakano Y, Hirano S, Kojima K, Li H, Sakurai T, Suzuki M, Tai H, Furukawa S, Sugiyama A, Yamanaka Y, Yamamoto T, Iimori T, Yokota H, Mukai H, Horikoshi T, Uno T, Kuwabara S. Dopaminergic Correlates of Regional Cerebral Blood Flow in Parkinsonian Disorders. Mov Disord 2022; 37:1235-1244. [DOI: 10.1002/mds.28981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yoshikazu Nakano
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
- Department of Neurology Chibaken Saiseikai Narashino Hospital Narashino Japan
| | - Shigeki Hirano
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Kazuho Kojima
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
- Department of Neurology Chiba Rosai Hospital Ichihara Japan
| | - Honglinag Li
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Toru Sakurai
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Masahide Suzuki
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Hong Tai
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Shogo Furukawa
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
- Department of Neurology Japanese Red Cross Narita Hospital Narita Japan
| | - Atsuhiko Sugiyama
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Yoshitaka Yamanaka
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| | - Tatsuya Yamamoto
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
- Division of Occupational Therapy, Department of Rehabilitation Chiba Prefectural University of Health Sciences Chiba Japan
| | - Takashi Iimori
- Department of Radiology Chiba University Hospital Chiba Japan
| | - Hajime Yokota
- Diagnostic Radiology and Radiation Oncology Graduate School of Medicine, Chiba University Chiba Japan
| | - Hiroki Mukai
- Department of Radiology Chiba University Hospital Chiba Japan
| | | | - Takashi Uno
- Diagnostic Radiology and Radiation Oncology Graduate School of Medicine, Chiba University Chiba Japan
| | - Satoshi Kuwabara
- Department of Neurology Graduate School of Medicine, Chiba University Chiba Japan
| |
Collapse
|
12
|
Vore AS, Deak T. Alcohol, inflammation, and blood-brain barrier function in health and disease across development. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:209-249. [PMID: 34801170 DOI: 10.1016/bs.irn.2021.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alcohol is the most commonly used drug of abuse in the world and binge drinking is especially harmful to the brain, though the mechanisms by which alcohol compromises overall brain health remain somewhat elusive. A number of brain diseases and pathological states are accompanied by perturbations in Blood-Brain Barrier (BBB) function, ultimately exacerbating disease progression. The BBB is critical for coordinating activity between the peripheral immune system and the brain. Importantly, BBB integrity is responsive to circulating cytokines and other immune-related signaling molecules, which are powerfully modulated by alcohol exposure. This review will highlight key cellular components of the BBB; discuss mechanisms by which permeability is achieved; offer insight into methodological approaches for assessing BBB integrity; and forecast how alcohol-induced changes in the peripheral and central immune systems might influence BBB function in individuals with a history of binge drinking and ultimately Alcohol Use Disorders (AUD).
Collapse
Affiliation(s)
- A S Vore
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton, NY, United States
| | - T Deak
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton, NY, United States.
| |
Collapse
|
13
|
Lan G, Wang P, Chan RB, Liu Z, Yu Z, Liu X, Yang Y, Zhang J. Astrocytic VEGFA: An essential mediator in blood-brain-barrier disruption in Parkinson's disease. Glia 2021; 70:337-353. [PMID: 34713920 DOI: 10.1002/glia.24109] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 12/29/2022]
Abstract
The integrity of blood-brain-barrier (BBB) is essential for normal brain functions, synaptic remodeling, and angiogenesis. BBB disruption is a common pathology during Parkinson's disease (PD), and has been hypothesized to contribute to the progression of PD. However, the molecular mechanism of BBB disruption in PD needs further investigation. Here, A53T PD mouse and a 3-cell type in vitro BBB model were used to study the roles of α-synuclein (α-syn) in BBB disruption with the key results confirmed in the brains of PD patients obtained at autopsy. The A53T PD mouse studies showed that the expression of tight junction-related proteins decreased, along with increased vascular permeability and accumulation of oligomeric α-syn in activated astrocytes in the brain. The in vitro BBB model studies demonstrated that treatment with oligomeric α-syn, but not monomeric or fibrillar α-syn, resulted in significant disruption of BBB integrity. This process involved the expression and release of vascular endothelial growth factor A (VEGFA) and nitric oxide (NO) from oligomeric α-syn treated astrocytes. Increased levels of VEGFA and iNOS were also observed in the brain of PD patients. Blocking the VEGFA signaling pathway in the in vitro BBB model effectively protected the barrier against the harmful effects of oligomeric α-syn. Finally, the protective effects on BBB integrity associated with inhibition of VEGFA signaling pathway was also confirmed in PD mice. Taken together, our study concluded that oligomeric α-syn is critically involved in PD-associated BBB disruption, in a process that is mediated by astrocyte-derived VEGFA.
Collapse
Affiliation(s)
- Guoyu Lan
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Pan Wang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang, China.,National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, Zhejiang, China
| | | | - Zongran Liu
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaodan Liu
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Ying Yang
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Jing Zhang
- Department of Pathology, Zhejiang University First Affiliated Hospital and School of Medicine, Hangzhou, Zhejiang, China.,National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Bidesi NSR, Vang Andersen I, Windhorst AD, Shalgunov V, Herth MM. The role of neuroimaging in Parkinson's disease. J Neurochem 2021; 159:660-689. [PMID: 34532856 PMCID: PMC9291628 DOI: 10.1111/jnc.15516] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects millions of people worldwide. Two hallmarks of PD are the accumulation of alpha-synuclein and the loss of dopaminergic neurons in the brain. There is no cure for PD, and all existing treatments focus on alleviating the symptoms. PD diagnosis is also based on the symptoms, such as abnormalities of movement, mood, and cognition observed in the patients. Molecular imaging methods such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), and positron emission tomography (PET) can detect objective alterations in the neurochemical machinery of the brain and help diagnose and study neurodegenerative diseases. This review addresses the application of functional MRI, PET, and SPECT in PD patients. We provide an overview of the imaging targets, discuss the rationale behind target selection, the agents (tracers) with which the imaging can be performed, and the main findings regarding each target's state in PD. Molecular imaging has proven itself effective in supporting clinical diagnosis of PD and has helped reveal that PD is a heterogeneous disorder, which has important implications for the development of future therapies. However, the application of molecular imaging for early diagnosis of PD or for differentiation between PD and atypical parkinsonisms has remained challenging. The final section of the review is dedicated to new imaging targets with which one can detect the PD-related pathological changes upstream from dopaminergic degeneration. The foremost of those targets is alpha-synuclein. We discuss the progress of tracer development achieved so far and challenges on the path toward alpha-synuclein imaging in humans.
Collapse
Affiliation(s)
- Natasha S R Bidesi
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Albert D Windhorst
- Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
15
|
Fujita K, Peng S, Ma Y, Tang CC, Hellman M, Feigin A, Eidelberg D, Dhawan V. Blood-brain barrier permeability in Parkinson's disease patients with and without dyskinesia. J Neurol 2021; 268:2246-2255. [PMID: 33502551 PMCID: PMC11197155 DOI: 10.1007/s00415-021-10411-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/09/2020] [Accepted: 01/15/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Recent studies on a rodent model of Parkinson's disease (PD) have raised the possibility of increased blood-brain barrier (BBB) permeability, demonstrated by histology, autoradiography, and positron emission tomography (PET). However, in human PD patients, in vivo evidence of increased BBB permeability is lacking. We examined the hypothesis that levodopa treatment increases BBB permeability in human subjects with PD, particularly in those with levodopa-induced dyskinesia (LID). METHODS We used rubidium-82 (82Rb) and PET to quantify BBB influx in vivo in 19 PD patients, including eight with LID, and 12 age- and sex-matched healthy subjects. All subjects underwent baseline 82Rb scans. Seventeen chronically levodopa-treated patients were additionally rescanned during intravenous levodopa infusion. Influx rate constant, K1, by compartmental modeling or net influx transport, Ki, by graphical approach could not be estimated reliably. However, Vd, the "apparent volume of distribution" based on the 82Rb concentration in brain tissue and blood, was estimated with good stability as a local measure of the volume of distribution. RESULTS Rubidium influx into brain tissue was undetectable in PD patients with or without LID, scanned on and off drug. No significant differences in regional Vd were observed for PD patients with or without LID relative to healthy subjects, except in left thalamus. Moreover, changes in Vd measured off- and on-levodopa infusion were also not significant for dyskinetic and non-dyskinetic subjects. CONCLUSION 82Rb PET did not reveal significant changes in BBB permeability in PD patients.
Collapse
Affiliation(s)
- Koji Fujita
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shichun Peng
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Yilong Ma
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Matthew Hellman
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Andrew Feigin
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Vijay Dhawan
- Center for Neurosciences, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
16
|
Sidtis JJ, Sidtis DVL, Dhawan V, Tagliati M, Eidelberg D. Stimulation of the Subthalamic Nucleus Changes Cortical-Subcortical Blood Flow Patterns During Speech: A Positron Emission Tomography Study. Front Neurol 2021; 12:684596. [PMID: 34122323 PMCID: PMC8187801 DOI: 10.3389/fneur.2021.684596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/05/2021] [Indexed: 12/18/2022] Open
Abstract
Background: Deep brain stimulation of the subthalamic nucleus (STN-DBS) is an effective treatment for Parkinson's disease (PD) but can have an adverse effect on speech. In normal speakers and in those with spinocerebellar ataxia, an inverse relationship between regional cerebral blood flow (rCBF) in the left inferior frontal (IFG) region and the right caudate (CAU) is associated with speech rate. This pattern was examined to determine if it was present in PD, and if so, whether it was altered by STN-DBS. Methods: Positron Emission Tomography (PET) measured rCBF during speech in individuals with PD not treated with STN-DBS (n = 7), and those treated with bilateral STN-DBS (n = 7). Previously reported results from non-PD control subjects (n = 16) were reported for comparison. The possible relationships between speech rate during scanning and data from the left and right IFG and CAU head regions were investigated using a step-wise multiple linear regression to identify brain regions that interacted to predict speech rate. Results: The multiple linear regression analysis replicated previously reported predictive coefficients for speech rate involving the left IFG and right CAU regions. However, the relationships between these predictive coefficients and speech rates were abnormal in both PD groups. In PD who had not received STN-DBS, the right CAU coefficient decreased normally with increasing speech rate but the left IFG coefficient abnormally decreased. With STN-DBS, this pattern was partially normalized with the addition of a left IFG coefficient that increased with speech rate, as in normal controls, but the abnormal left IFG decreasing coefficient observed in PD remained. The magnitudes of both cortical predictive coefficients but not the CAU coefficient were exaggerated with STN-DBS. Conclusions: STN-DBS partially corrects the abnormal relationships between rCBF and speech rate found in PD by introducing a left IFG subregion that increases with speech rate, but the conflicting left IFG subregion response remained. Conflicting IFG responses may account for some of the speech problems observed after STN-DBS. Cortical and subcortical regions may be differentially affected by STN-DBS.
Collapse
Affiliation(s)
- John J Sidtis
- Brain and Behavior Laboratory, Geriatrics Department, Nathan Kline Institute, Orangeburg, NY, United States.,Department of Psychiatry, School of Medicine, New York University Langone, New York, NY, United States
| | - Diana Van Lancker Sidtis
- Brain and Behavior Laboratory, Geriatrics Department, Nathan Kline Institute, Orangeburg, NY, United States.,Department of Communicative Disorders and Sciences, New York University Steinhardt School, New York, NY, United States
| | - Vijay Dhawan
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Michele Tagliati
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
17
|
Booth S, Ramadan A, Zhang D, Lu L, Kirouac G, Jackson MF, Anderson C, Ko JH. The Vasomotor Response to Dopamine Is Altered in the Rat Model of l-dopa-Induced Dyskinesia. Mov Disord 2021; 36:938-947. [PMID: 33135810 PMCID: PMC8246949 DOI: 10.1002/mds.28357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/08/2020] [Accepted: 10/01/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Levodopa (l-dopa) is the frontline treatment for motor symptoms of Parkinson's disease. However, prolonged use of l-dopa results in a motor complication known as levodopa-induced dyskinesia (LID) in ~50% of patients over 5 years. OBJECTIVES We investigated neurovascular abnormalities in a rat model of LID by examining changes in angiogenesis and dopamine-dependent vessel diameter changes. METHODS Differences in striatal and nigral angiogenesis in a parkinsonian rat model (6-OHDA lesion) treated with 2 doses of l-dopa (saline, 2, and 10 mg/kg/day subcutaneous l-dopa treatment for 22 days) by 5-bromo-2'-deoxyuridine (BrdU)-RECA1 co-immunofluorescence. Difference in the vasomotor response to dopamine was examined with 2-photon laser scanning microscopy and Dodt gradient imaging. RESULTS We found that the 10 mg/kg l-dopa dosing regimen induced LID in all animals (n = 5) and induced significant angiogenesis in the striatum and substantia nigra. In contrast, the 2 mg/kg treatment induced LID in 6 out of 12 rats and led to linearly increasing LID severity over the 22-day treatment period, making this a promising model for studying LID progression longitudinally. However, no significantly different level of angiogenesis was observed between LID versus non-LID animals. Dopamine-induced vasodilatory responses were exaggerated only in rats that show LID-like signs compared to the rest of groups. Additionally, in juvenile rats, we showed that DA-induced vasodilation is preceded by increased Ca2+ release in the adjacent astrocytes. CONCLUSION This finding supports that astrocytic dopamine signaling controls striatal blood flow bidirectionally, and the balance is altered in LID. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Samuel Booth
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegManitobaCanada
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
| | - Abdullah Ramadan
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegManitobaCanada
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
| | - Dali Zhang
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegManitobaCanada
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
| | - Lingling Lu
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
- Department of Pharmacology and TherapeuticsUniversity of ManitobaWinnipegManitobaCanada
| | - Gilbert Kirouac
- Department of Oral BiologyUniversity of ManitobaWinnipegManitobaCanada
| | - Michael F. Jackson
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
- Department of Pharmacology and TherapeuticsUniversity of ManitobaWinnipegManitobaCanada
| | - Chris Anderson
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
- Department of Pharmacology and TherapeuticsUniversity of ManitobaWinnipegManitobaCanada
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell ScienceUniversity of ManitobaWinnipegManitobaCanada
- Kleyson Institute for Advanced MedicineHealth Science CentreWinnipegManitobaCanada
| |
Collapse
|
18
|
Peng S, Tang C, Schindlbeck K, Rydzinski Y, Dhawan V, Spetsieris PG, Ma Y, Eidelberg D. Dynamic 18F-FPCIT PET: Quantification of Parkinson's disease metabolic networks and nigrostriatal dopaminergic dysfunction in a single imaging session. J Nucl Med 2021; 62:jnumed.120.257345. [PMID: 33741649 PMCID: PMC8612203 DOI: 10.2967/jnumed.120.257345] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Previous multi-center imaging studies with 18F-FDG PET have established the presence of Parkinson's disease motor- and cognition-related metabolic patterns termed PDRP and PDCP in patients with this disorder. Given that in PD cerebral perfusion and glucose metabolism are typically coupled in the absence of medication, we determined whether subject expression of these disease networks can be quantified in early-phase images from dynamic 18F-FPCIT PET scans acquired to assess striatal dopamine transporter (DAT) binding. Methods: We studied a cohort of early-stage PD patients and age-matched healthy control subjects who underwent 18F-FPCIT at baseline; scans were repeated 4 years later in a smaller subset of patients. The early 18F-FPCIT frames, which reflect cerebral perfusion, were used to compute PDRP and PDCP expression (subject scores) in each subject, and compared to analogous measures computed based on 18F-FDG PET scan when additionally available. The late 18F-FPCIT frames were used to measure caudate and putamen DAT binding in the same individuals. Results: PDRP subject scores from early-phase 18F-FPCIT and 18F-FDG scans were elevated and striatal DAT binding reduced in PD versus healthy subjects. The PDRP scores from 18F-FPCIT correlated with clinical motor ratings, disease duration, and with corresponding measures from 18F-FDG PET. In addition to correlating with disease duration and analogous 18F-FDG PET values, PDCP scores correlated with DAT binding in the caudate/anterior putamen. PDRP and PDCP subject scores using either method rose over 4 years whereas striatal DAT binding declined over the same time period. Conclusion: Early-phase images obtained with 18F-FPCIT PET can provide an alternative to 18F-FDG PET for PD network quantification. This technique therefore allows PDRP/PDCP expression and caudate/putamen DAT binding to be evaluated with a single tracer in one scanning session.
Collapse
Affiliation(s)
- Shichun Peng
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - Chris Tang
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - Katharina Schindlbeck
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - Yaacov Rydzinski
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vijay Dhawan
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - Phoebe G. Spetsieris
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - Yilong Ma
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| | - David Eidelberg
- Center for Neurosciences, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York; and
| |
Collapse
|
19
|
Elevated caudate connectivity in cognitively normal Parkinson's disease patients. Sci Rep 2020; 10:17978. [PMID: 33087833 PMCID: PMC7578639 DOI: 10.1038/s41598-020-75008-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 09/29/2020] [Indexed: 01/29/2023] Open
Abstract
Mild cognitive impairment (MCI) is common in Parkinson’s disease patients. However, its underlying mechanism is not well understood, which has hindered new treatment discoveries specific to MCI. The aim of this study was to investigate functional connectivity changes of the caudate nucleus in cognitively impaired Parkinson’s patients. We recruited 18 Parkinson’s disease patients—10 PDNC [normal cognition Parkinson’s disease; Montreal Cognitive Assessment (MoCA) ≥ 26], 8 PDLC (low cognition Parkinson’s disease; MoCA < 26) —and 10 age-matched healthy controls. All subjects were scanned with resting-state functional magnetic resonance imaging (MRI) and perfusion MRI. We analyzed these data for graph theory metrics and Alzheimer’s disease-like pattern score, respectively. A strong positive correlation was found between the functional connectivity of the right caudate nucleus and MoCA scores in Parkinson’s patient groups, but not in healthy control subjects. Interestingly, PDNC’s functional connectivity of the right caudate was significantly higher than both PDLC and healthy controls, while PDLC and healthy controls were not significantly different from each other. We found that Alzheimer’s disease-like metabolic/perfusion pattern score correlated with MoCA scores in healthy controls, but not in Parkinson’s disease. Increased caudate connectivity may be related to a compensatory mechanism found in cognitively normal patients with Parkinson’s disease. Our findings support and complement the dual syndrome hypothesis.
Collapse
|
20
|
Fletcher EJR, Finlay CJ, Amor Lopez A, Crum WR, Vernon AC, Duty S. Neuroanatomical and Microglial Alterations in the Striatum of Levodopa-Treated, Dyskinetic Hemi-Parkinsonian Rats. Front Neurosci 2020; 14:567222. [PMID: 33041762 PMCID: PMC7522511 DOI: 10.3389/fnins.2020.567222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/17/2020] [Indexed: 02/02/2023] Open
Abstract
Dyskinesia associated with chronic levodopa treatment in Parkinson’s disease is associated with maladaptive striatal plasticity. The objective of this study was to examine whether macroscale structural changes, as captured by magnetic resonance imaging (MRI) accompany this plasticity and to identify plausible cellular contributors in a rodent model of levodopa-induced dyskinesia. Adult male Sprague-Dawley rats were rendered hemi-parkinsonian by stereotaxic injection of 6-hydroxydopamine into the left medial forebrain bundle prior to chronic treatment with saline (control) or levodopa to induce abnormal involuntary movements (AIMs), reflective of dyskinesia. Perfusion-fixed brains underwent ex vivo structural MRI before sectioning and staining for cellular markers. Chronic treatment with levodopa induced significant AIMs (p < 0.0001 versus saline). The absolute volume of the ipsilateral, lesioned striatum was increased in levodopa-treated rats resulting in a significant difference in percentage volume change when compared to saline-treated rats (p < 0.01). Moreover, a significant positive correlation was found between this volume change and AIMs scores for individual levodopa-treated rats (r = 0.96; p < 0.01). The density of Iba1+ cells was increased within the lesioned versus intact striatum (p < 0.01) with no difference between treatment groups. Conversely, Iba1+ microglia soma size was significantly increased (p < 0.01) in the lesioned striatum of levodopa-treated but not saline-treated rats. Soma size was not, however, significantly correlated with either AIMs or MRI volume change. Although GFAP+ astrocytes were elevated in the lesioned versus intact striatum (p < 0.001), there was no difference between treatment groups. No statistically significant effects of either lesion or treatment on RECA1, a marker for blood vessels, were observed. Collectively, these data suggest chronic levodopa treatment in 6-hydroxydopamine lesioned rats is associated with increased striatal volume that correlates with the development of AIMs. The accompanying increase in number and size of microglia, however, cannot alone explain this volume expansion. Further multi-modal studies are warranted to establish the brain-wide effects of chronic levodopa treatment.
Collapse
Affiliation(s)
- Edward J R Fletcher
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Clare J Finlay
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ana Amor Lopez
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - William R Crum
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Susan Duty
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
21
|
Falquetto B, Thieme K, Malta MB, e Rocha KC, Tuppy M, Potje SR, Antoniali C, Rodrigues AC, Munhoz CD, Moreira TS, Takakura AC. Oxidative stress in the medullary respiratory neurons contributes to respiratory dysfunction in the 6‐OHDA model of Parkinson's disease. J Physiol 2020; 598:5271-5293. [DOI: 10.1113/jp279791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/14/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Bárbara Falquetto
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Karina Thieme
- Department of Physiology and Biophysics Instituto de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Marília B. Malta
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Karina C. e Rocha
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Marina Tuppy
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Simone R. Potje
- Department of Basic Sciences School of Dentistry São Paulo State University (UNESP) Araçatuba SP 16015‐050 Brazil
| | - Cristina Antoniali
- Department of Basic Sciences School of Dentistry São Paulo State University (UNESP) Araçatuba SP 16015‐050 Brazil
| | - Alice C. Rodrigues
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Carolina D. Munhoz
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Thiago S. Moreira
- Department of Physiology and Biophysics Instituto de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| | - Ana C. Takakura
- Department of Pharmacology Institute de Ciencias Biomedicas Universidade de Sao Paulo São Paulo SP 05508‐000 Brazil
| |
Collapse
|
22
|
Xing Y, Tench C, Wongwandee M, Schwarz ST, Bajaj N, Auer DP. Coordinate based meta-analysis of motor functional imaging in Parkinson's: disease-specific patterns and modulation by dopamine replacement and deep brain stimulation. Brain Imaging Behav 2020; 14:1263-1280. [PMID: 30809759 PMCID: PMC7381438 DOI: 10.1007/s11682-019-00061-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate factors affecting the pattern of motor brain activation reported in people with Parkinson's (PwP), aiming to differentiate disease-specific features from treatment effects. METHODS A co-ordinate-based-meta-analysis (CBMA) of functional motor neuroimaging studies involving patients with Parkinson's (PwP), and healthy controls (HC) identified 126 suitable articles. The experiments were grouped based on subject feature, medication status (onMed/offMed), deep brain stimulation (DBS) status (DBSon/DBSoff) and type of motor initiation. RESULTS HC and PwP shared similar neural networks during upper extremity motor tasks but with differences of reported frequency in mainly bilateral putamen, insula and ipsilateral inferior parietal and precentral gyri. The activation height was significantly reduced in the bilateral putamen, left SMA, left subthalamus nucleus, right thalamus and right midial global pallidum in PwPoffMed (vs. HC), and pre-SMA hypoactivation correlated with disease severity. These changes were not found in patients on dopamine replacement therapy (PwPonMed vs. HC) in line with a restorative function. By contrast, left SMA and primary motor cortex showed hyperactivation in the medicated state (vs. HC) suggesting dopaminergic overcompensation. Deep-brain stimulation (PwP during the high frequency subthalamus nucleus (STN) DBS vs. no stimulation) induced a decrease in left SMA activity and the expected increase in the left subthalamic/thalamic region regardless of hand movement. We further demonstrated a disease related effect of motor intention with only PwPoffMed showing increased activation in the medial frontal lobe in self-initiated studies. CONCLUSION We describe a consistent disease-specific pattern of putaminal hypoactivation during motor tasks that appears reversed by dopamine replacement. Inconsistent reports of altered SMA/pre-SMA activation can be explained by task- and medication-specific variation in intention. Moreover, SMA activity was reduced during STN-DBS, while dopamine-induced hyperactivation of SMA which might underpin hyperdynamic L-dopa related overcompensation.
Collapse
Affiliation(s)
- Yue Xing
- Radiological Sciences, Division of Clinical Neuroscience, University of Nottingham, Nottingham, NG7 2UH, UK.
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
- Radiological Sciences, Sir Peter Mansfield Imaging Centre, NIHR Nottingham BRC, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - Christopher Tench
- Division of Clinical Neurology, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Monton Wongwandee
- Department of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Stefan T Schwarz
- Radiological Sciences, Division of Clinical Neuroscience, University of Nottingham, Nottingham, NG7 2UH, UK
- Department of Radiology, Cardiff and Vale University Health Board, Cardiff, Wales
| | - Nin Bajaj
- Department of Neurology, Nottingham University Hospitals, Nottingham, NG7 2UH, UK
| | - Dorothee P Auer
- Radiological Sciences, Division of Clinical Neuroscience, University of Nottingham, Nottingham, NG7 2UH, UK.
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
- Radiological Sciences, Sir Peter Mansfield Imaging Centre, NIHR Nottingham BRC, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
23
|
Regulation of immune-driven pathogenesis in Parkinson's disease by gut microbiota. Brain Behav Immun 2020; 87:890-897. [PMID: 31931152 DOI: 10.1016/j.bbi.2020.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is one of the most significant medical and social burdens of our time. The prevalence of PD increases with age and the number of individuals diagnosed with PD is expected to double from 6.9 million in 2015 to 14.2 million in 2040. To date, no drugs can stop the ongoing neurodegeneration caused by PD due to its unclear and complex pathogenic mechanisms. It has been wildly recognized that both gut microbiota and neuro-immunity are involved in the pathology of PD. In this review, we intend to provide a comprehensive overview of current knowledge on how gut microbiota involved in immune-driven pathogenesis of PD, and its potential as a new target of dietary and/or therapeutic interventions for PD.
Collapse
|
24
|
Rane S, Koh N, Oakley J, Caso C, Zabetian CP, Cholerton B, Montine TJ, Grabowski T. Arterial spin labeling detects perfusion patterns related to motor symptoms in Parkinson's disease. Parkinsonism Relat Disord 2020; 76:21-28. [PMID: 32559629 PMCID: PMC7554132 DOI: 10.1016/j.parkreldis.2020.05.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/25/2020] [Accepted: 05/08/2020] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Imaging neurovascular disturbances in Parkinson's disease (PD) is an excellent measure of disease severity. Indeed, a disease-specific regional pattern of abnormal metabolism has been identified using positron emission tomography. Only a handful of studies, however, have applied perfusion MRI to detect this disease pattern. Our goal was to replicate the evaluation of a PD-related perfusion pattern using scaled subprofile modeling/principal component analysis (SSM-PCA). METHODS We applied arterial spin labeling (ASL) MRI for this purpose. Uniquely, we assessed this pattern separately in PD individuals ON and OFF dopamine medications. We further compared the existence of these patterns and their strength in each individual with their Movement Disorder Society-Unified Parkinson's Disease Rating Scale motor (MDS-UPDRS) scores, cholinergic tone as indexed by short-term afferent inhibition (SAI), and other neuropsychiatric tests. RESULTS We observed a PD-related perfusion pattern that was similar to previous studies. The patterns were observed in both ON and OFF states but only the pattern in the OFF condition could significantly (AUC=0.72) differentiate between PD and healthy subjects. In the ON condition, PD subjects were similar to controls from a CBF standpoint (AUC=0.45). The OFF pattern prominently included the posterior cingulate, precentral region, precuneus, and the subcallosal cortex. Individual principal components from the ON and OFF states were strongly associated with MDS-UPDRS scores, SAI amplitude and latency. CONCLUSION Using ASL, our study identified patterns of abnormal perfusion in PD and were associated with disease symptoms.
Collapse
Affiliation(s)
- Swati Rane
- Integrated Brain Imaging Center, Radiology, University of Washington Medical Center, Seattle, WA, USA.
| | - Natalie Koh
- Integrated Brain Imaging Center, Radiology, University of Washington Medical Center, Seattle, WA, USA
| | - John Oakley
- Department of Neurology, University of Washington Medical Center, Seattle, WA, USA
| | - Christina Caso
- Integrated Brain Imaging Center, Radiology, University of Washington Medical Center, Seattle, WA, USA
| | - Cyrus P Zabetian
- Department of Neurology, University of Washington Medical Center, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Brenna Cholerton
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Thomas Grabowski
- Integrated Brain Imaging Center, Radiology, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
25
|
Liu C, Jiang J, Zhou H, Zhang H, Wang M, Jiang J, Wu P, Ge J, Wang J, Ma Y, Zuo C. Brain Functional and Structural Signatures in Parkinson's Disease. Front Aging Neurosci 2020; 12:125. [PMID: 32528272 PMCID: PMC7264099 DOI: 10.3389/fnagi.2020.00125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 04/14/2020] [Indexed: 11/13/2022] Open
Abstract
The aim of this study is to explore functional and structural properties of abnormal brain networks associated with Parkinson’s disease (PD). 18F-Fluorodeoxyglucose positron emission tomography (18F-FDG PET) and T1-weighted magnetic resonance imaging from 20 patients with moderate-stage PD and 20 age-matched healthy controls were acquired to identify disease-related patterns in functional and structural networks. Dual-modal images from another prospective subject of 15 PD patients were used as the validation group. Scaled Subprofile Modeling based on principal component analysis method was applied to determine disease-related patterns in both modalities, and brain connectome analysis based on graph theory was applied to verify these patterns. The results showed that the expressions of the metabolic and structural patterns in PD patients were significantly higher than healthy controls (PD1-HC, p = 0.0039, p = 0.0058; PD2-HC, p < 0.001, p = 0.044). The metabolic pattern was characterized by relative increased metabolic activity in pallidothalamic, pons, putamen, and cerebellum, associated with metabolic decreased in parietal–occipital areas. The structural pattern was characterized by relative decreased gray matter (GM) volume in pons, transverse temporal gyrus, left cuneus, right superior occipital gyrus, and right superior parietal lobule, associated with preservation in GM volume in pallidum and putamen. In addition, both patterns were verified in the connectome analysis. The findings suggest that significant overlaps between metabolic and structural patterns provide new evidence for elucidating the neuropathological mechanisms of PD.
Collapse
Affiliation(s)
- Chunhua Liu
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Jiehui Jiang
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,Key Laboratory of Specialty Fiber Optics and Optical Access Networks, Joint International Research Laboratory of Specialty Fiber Optics and Advanced Communication, Shanghai University, Shanghai, China
| | - Hucheng Zhou
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Huiwei Zhang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Wang
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Juanjuan Jiang
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingjie Ge
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yilong Ma
- Center for Neurosciences, Feinstein Institute for Medical Research, North Shore-Long Island Jewish Health System, Manhasset, NY, United States
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
27
|
Marín-Lahoz J, Sampedro F, Horta-Barba A, Martínez-Horta S, Aracil-Bolaños I, Camacho V, Bejr-kasem H, Pascual-Sedano B, Pérez-Pérez J, Gironell A, Pagonabarraga J, Carrió I, Kulisevsky J. Preservation of brain metabolism in recently diagnosed Parkinson’s impulse control disorders. Eur J Nucl Med Mol Imaging 2020; 47:2165-2174. [DOI: 10.1007/s00259-019-04664-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/16/2019] [Indexed: 12/16/2022]
|
28
|
Wei H, Zhou Y, Zhao J, Zhan L. Risk Factors and Metabolism of Different Brain Regions by Positron Emission Tomography in Parkinson Disease with Disabling Dyskinesia. Curr Neurovasc Res 2019; 16:310-320. [PMID: 31622205 DOI: 10.2174/1567202616666191009102112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/08/2019] [Accepted: 07/25/2019] [Indexed: 11/22/2022]
Abstract
Objective:Dyskinesia is the most common motor complication in advanced Parkinson’s Disease (PD) and has a severe impact on daily life. But the mechanism of dyskinesia is still poorly understood. This study aims to explore risk factors for disabling dyskinesia in PD and further analyze the Vesicular Monoamine Transporter 2 (VMAT2) distribution (labeled with 18F-AV133) in the corpus striatum and the 18F-fluorodeoxyglucose (18F-FDG) metabolism of different brain regions by PET-CT.Methods:This is a cross-sectional study involving 135 PD patients. They were divided into disabling dyskinesia group (DD group, N=22) and non-dyskinesia group (ND group, N=113). All the patients were agreed to undergo PET-CT scans. Clinical data were analyzed between two groups by using multivariate logistic regression analysis, and risk factors for disabling dyskinesia were then determined. The standard uptake value ratios (SUVr) of 18F-AV133 in the corpus striatum and the 18F-FDG metabolism of different brain regions were identified and calculated by the software.Results:6.3% patients have disabling dyskinesia. DD group were more likely to have longer Disease Duration, higher Hoehn-Yahr degree, more severe clinic symptoms, more frequent sleep behavior disorder, and higher levodopa dose equivalency than ND group (P < 0.05). After adjusting confounding factors by multivariate logistic regression, DD group had longer PD duration and high levodopa dose equivalency compared with ND group (P < 0.05). There is no significant difference between the VMAT2 distribution (labeled with 18F- AV133) in the putamen and caudate between two groups. And the 18F-FDG metabolic changes in cortical and subcortical regions did not show a significant difference between the two groups either (P > 0.05).Conclusion:Long PD duration and high levodopa dose equivalency were two independent risk factors for disabling dyskinesia in PD patients. Compared to non-dyskinesia PD patients, there was no significant dopamine decline of the nigrostriatal system in disabling dyskinesia PD patients. Activities of different brain regions were not different between the two groups by 18F-FDG PETCT.
Collapse
Affiliation(s)
- Huan Wei
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yongtao Zhou
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Junwu Zhao
- Department of Neurology, Weihai Municipal Hospital, Shandong, China
| | - Liping Zhan
- Department of Neurology, The Affiliated Yan'an Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
29
|
Boi L, Pisanu A, Greig NH, Scerba MT, Tweedie D, Mulas G, Fenu S, Carboni E, Spiga S, Carta AR. Immunomodulatory drugs alleviate l-dopa-induced dyskinesia in a rat model of Parkinson's disease. Mov Disord 2019; 34:1818-1830. [PMID: 31335998 PMCID: PMC11719776 DOI: 10.1002/mds.27799] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/05/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Thalidomide and closely related analogues are used clinically for their immunomodulatory and antiangiogenic properties mediated by the inhibition of the proinflammatory cytokine tumor necrosis factor α. Neuroinflammation and angiogenesis contribute to classical neuronal mechanisms underpinning the pathophysiology of l-dopa-induced dyskinesia, a motor complication associated with l-dopa therapy in Parkinson's disease. The efficacy of thalidomide and the more potent derivative 3,6'-dithiothalidomide on dyskinesia was tested in the 6-hydroxydopamine Parkinson's disease model. METHODS Three weeks after 6-hydroxydopamine infusion, rats received 10 days of treatment with l-dopa plus benserazide (6 mg/kg each) and thalidomide (70 mg/kg) or 3,6'-dithiothalidomide (56 mg/kg), and dyskinesia and contralateral turning were recorded daily. Rats were euthanized 1 hour after the last l-dopa injection, and levels of tumor necrosis factor-α, interleukin-10, OX-42, vimentin, and vascular endothelial growth factor immunoreactivity were measured in their striatum and substantia nigra reticulata to evaluate neuroinflammation and angiogenesis. Striatal levels of GLUR1 were measured as a l-dopa-induced postsynaptic change that is under tumor necrosis factor-α control. RESULTS Thalidomide and 3,6'-dithiothalidomide significantly attenuated the severity of l-dopa-induced dyskinesia while not affecting contralateral turning. Moreover, both compounds inhibited the l-dopa-induced microgliosis and excessive tumor necrosis factor-α in the striatum and substantia nigra reticulata, while restoring physiological levels of the anti-inflammatory cytokine interleukin-10. l-Dopa-induced angiogenesis was inhibited in both basal ganglia nuclei, and l-dopa-induced GLUR1 overexpression in the dorsolateral striatum was restored to normal levels. CONCLUSIONS These data suggest that decreasing tumor necrosis factor-α levels may be useful to reduce the appearance of dyskinesia, and thalidomide, and more potent derivatives may provide an effective therapeutic approach to dyskinesia. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Laura Boi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Augusta Pisanu
- CNR Institute of Neuroscience, Cagliari, Cagliari, Italy
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Michael T. Scerba
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, National Institute of Aging, Baltimore, Maryland, USA
| | - Giovanna Mulas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Sandro Fenu
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Saturnino Spiga
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- National Institute of Neuroscience (INN), University of Cagliari, Cagliari, Italy
| |
Collapse
|
30
|
Aljuaid M, Booth S, Hobson DE, Borys A, Williams K, Katako A, Ryner L, Goertzen AL, Ko JH. Blood Flow and Glucose Metabolism Dissociation in the Putamen Is Predictive of Levodopa Induced Dyskinesia in Parkinson's Disease Patients. Front Neurol 2019; 10:1217. [PMID: 31824400 PMCID: PMC6881455 DOI: 10.3389/fneur.2019.01217] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/01/2019] [Indexed: 12/28/2022] Open
Abstract
Background: The forefront treatment of Parkinson's disease (PD) is Levodopa. When patients are treated with Levodopa cerebral blood flow is increased while cerebral metabolic rate is decreased in key subcortical regions including the putamen. This phenomenon is especially pronounced in patients with Levodopa-induced dyskinesia (LID). Method: To study the effect of clinically-determined anti-parkinsonian medications, 10 PD patients (5 with LID and 5 without LID) have been scanned with FDG-PET (a probe for glucose metabolism) and perfusion MRI (a probe for cerebral blood flow) both when they are ON and OFF medications. Patients additionally underwent resting state fMRI to detect changes in dopamine-mediated cortico-striatal connectivity. The degree of blood flow-glucose metabolism dissociation was quantified by comparing the FDG-PET and perfusion MRI data. Results: A significant interaction effect (imaging modality × medication; blood flow-glucose metabolism dissociation) has been found in the putamen (p = 0.023). Post-hoc analysis revealed that anti-parkinsonian medication consistently normalized the pathologically hyper-metabolic state of the putamen while mixed effects were observed in cerebral blood flow changes. This dissociation was especially predominant in patients with LID compared to those without. Unlike the prior study, this differentiation was not observed when cortico-striatal functional connectivity was assessed. Conclusion: We confirmed striatal neurovascular dissociation between FDG-PET and perfusion MRI in response to clinically determined anti-parkinsonian medication. We further proposed a novel analytical method to quantify the degree of dissociation in the putamen using only the ON condition scans, Putamen-to-thalamus Hyper-perfusion/hypo-metabolism Index (PHI), which may have the potential to be used as a biomarker for LID (correctly classifying 8 out 10 patients). For wider use of PHI, a larger validation study is warranted.
Collapse
Affiliation(s)
- Maram Aljuaid
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Samuel Booth
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Douglas E Hobson
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew Borys
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Kelly Williams
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Audrey Katako
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Lawrence Ryner
- Department of Radiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew L Goertzen
- Department of Radiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| |
Collapse
|
31
|
Ko JH, Spetsieris PG, Eidelberg D. Network Structure and Function in Parkinson's Disease. Cereb Cortex 2019; 28:4121-4135. [PMID: 29088324 DOI: 10.1093/cercor/bhx267] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Little is known of the structural and functional properties of abnormal brain networks associated with neurological disorders. We used a social network approach to characterize the properties of the Parkinson's disease (PD) metabolic topography in 4 independent patient samples and in an experimental non-human primate model. The PD network exhibited distinct features. Dense, mutually facilitating functional connections linked the putamen, globus pallidus, and thalamus to form a metabolically active core. The periphery was formed by weaker connections linking less active cortical regions. Notably, the network contained a separate module defined by interconnected, metabolically active nodes in the cerebellum, pons, frontal cortex, and limbic regions. Exaggeration of the small-world property was a consistent feature of disease networks in parkinsonian humans and in the non-human primate model; this abnormality was only partly corrected by dopaminergic treatment. The findings point to disease-related alterations in network structure and function as the basis for faulty information processing in this disorder.
Collapse
Affiliation(s)
- Ji Hyun Ko
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Phoebe G Spetsieris
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, USA.,Department of Neurology, Northwell Health, Manhasset, NY, USA
| |
Collapse
|
32
|
Xie J, Shen Z, Anraku Y, Kataoka K, Chen X. Nanomaterial-based blood-brain-barrier (BBB) crossing strategies. Biomaterials 2019; 224:119491. [PMID: 31546096 DOI: 10.1016/j.biomaterials.2019.119491] [Citation(s) in RCA: 339] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/31/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022]
Abstract
Increasing attention has been paid to the diseases of central nervous system (CNS). The penetration efficiency of most CNS drugs into the brain parenchyma is rather limited due to the existence of blood-brain barrier (BBB). Thus, BBB crossing for drug delivery to CNS remains a significant challenge in the development of neurological therapeutics. Because of the advantageous properties (e.g., relatively high drug loading content, controllable drug release, excellent passive and active targeting, good stability, biodegradability, biocompatibility, and low toxicity), nanomaterials with BBB-crossability have been widely developed for the treatment of CNS diseases. This review summarizes the current understanding of the physiological structure of BBB, and provides various nanomaterial-based BBB-crossing strategies for brain delivery of theranostic agents, including intranasal delivery, temporary disruption of BBB, local delivery, cell penetrating peptide (CPP) mediated BBB-crossing, receptor mediated BBB-crossing, shuttle peptide mediated BBB-crossing, and cells mediated BBB-crossing. Clinicians, biologists, material scientists and chemists are expected to be interested in this review.
Collapse
Affiliation(s)
- Jinbing Xie
- Jiangsu Key Laboratory of Molecular Imaging and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China; Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Yasutaka Anraku
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan; Policy Alternatives Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
33
|
|
34
|
Motor Improvement-Related Regional Cerebral Blood Flow Changes in Parkinson's Disease in Response to Antiparkinsonian Drugs. PARKINSON'S DISEASE 2019; 2019:7503230. [PMID: 30944721 PMCID: PMC6421789 DOI: 10.1155/2019/7503230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/10/2019] [Accepted: 01/27/2019] [Indexed: 11/17/2022]
Abstract
Little is known about the relationship between regional cerebral blood flow (rCBF) change and clinical improvement in patients with Parkinson's disease (PD). Single-photon emission computed tomography (SPECT) measurement of cerebral blood flow allows evaluation of temporal changes in brain function, and using SPECT, we aimed to identify motor improvement-related rCBF changes in response to the administration of antiparkinsonian drugs. Thirty PD patients (16 without dementia; 14 with dementia) were scanned with technetium-99m labeled ethyl cysteinate dimer SPECT and were rated with the Movement Disorder Society-Unified Parkinson's Disease Rating Scale part III, both before and after a single administration of antiparkinsonian drugs. The SPECT data were processed using Statistical Parametric Mapping 2, the easy Z-score Imaging System, and voxel-based Stereotactic Extraction Estimation. The rCBF responses in the deep brain structures after administration of antiparkinsonian drugs tended to be larger than those in cortical areas. Among these deep brain structures, the rCBF increases in the substantia nigra (SN), lateral geniculate (LG) body, and medial geniculate (MG) body correlated with drug efficacy (p < 0.05, respectively). A subgroup analysis revealed that the motor improvement-related rCBF change in the MG was statistically significant, irrespective of cognitive function, but the significant changes in the LG and SN were not found in subjects with dementia. In conclusion, our SPECT study clearly exhibited drug-driven rCBF changes in PD patients, and we newly identified motor improvement-related rCBF changes in the LG and MG. These results suggest that rCBF changes in these regions could be considered as candidates for clinical indicators for objective evaluation of disease progression. Furthermore, functional studies focusing on the LG and MG, especially in relation to therapies using audio-visual stimuli, may bring some new clues to explain the pathophysiology of PD.
Collapse
|
35
|
Pathophysiology of levodopa-induced dyskinesia: Insights from multimodal imaging and immunohistochemistry in non-human primates. Neuroimage 2018; 183:132-141. [DOI: 10.1016/j.neuroimage.2018.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/21/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022] Open
|
36
|
Sharif Y, Jumah F, Coplan L, Krosser A, Sharif K, Tubbs RS. Blood brain barrier: A review of its anatomy and physiology in health and disease. Clin Anat 2018; 31:812-823. [PMID: 29637627 DOI: 10.1002/ca.23083] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/03/2018] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is the principal regulator of transport of molecules and cells into and out of the central nervous system (CNS). It comprises endothelial cells, pericytes, immune cells, astrocytes, and basement membrane, collectively known as the neurovascular unit. The development of the barrier involves many complex pathways from all the progenitors of the neurovascular unit, but the timing of its formation is not entirely known. The coordinated activities of all the components of the neurovascular unit and other tissues ensure that materials required for growth and maintenance are allowed into the CNS while extraneous ones are excluded. This review summarizes current knowledge of the anatomy, development, and physiology of the BBB, and alterations that occur in disease conditions. Clin. Anat. 31:812-823, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yousra Sharif
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Fareed Jumah
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Louis Coplan
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alec Krosser
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kassem Sharif
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - R Shane Tubbs
- Department of Anatomical Sciences, St. George's University, Grenada.,Seattle Science Foundation, Seattle, Washington
| |
Collapse
|
37
|
Kübel S, Stegmayer K, Vanbellingen T, Walther S, Bohlhalter S. Deficient supplementary motor area at rest: Neural basis of limb kinetic deficits in Parkinson's disease. Hum Brain Mapp 2018; 39:3691-3700. [PMID: 29722099 DOI: 10.1002/hbm.24204] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/16/2018] [Accepted: 04/23/2018] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) patients frequently suffer from limb kinetic apraxia (LKA) affecting quality of life. LKA denotes an impairment of precise and independent finger movements beyond bradykinesia, which is reliably assessed by coin rotation (CR) task. BOLD fMRI detected activation of a left inferior parietal-premotor praxis network in PD during CR. Here, we explored which network site is most critical for LKA using arterial spin labeling (ASL). Based on a hierarchical model, we hypothesized that LKA would predominantly affect the functional integrity of premotor areas including supplementary motor areas (SMA). Furthermore, we suspected that for praxis function with higher demand on temporal-spatial processing such as gesturing, inferior parietal lobule (IPL) upstream to premotor areas would be essential. A total of 21 PD patients and 20 healthy controls underwent ASL acquisition during rest. Behavioral assessment outside the scanner involved the CR, finger tapping task, and the test of upper limb apraxia (TULIA). Whole-brain analysis of activity at rest showed a significant reduction of CR-related perfusion in the left SMA of PD. Furthermore, the positive correlation between SMA perfusion and CR, seen in controls, was lost in patients. By contrast, TULIA was significantly associated with the perfusion of left IPL in both patients and controls. In conclusion, the findings suggest that LKA in PD are linked to an intrinsic disruption of the left SMA function, which may only be overcome by compensatory network activation. In addition, gestural performance relies on IPL which remains available for functional recruitment in early PD.
Collapse
Affiliation(s)
- Stefanie Kübel
- Neurocenter, Luzerner Kantonsspital, Spitalstrasse 31, Luzern 16, 6000, Switzerland
| | - Katharina Stegmayer
- University Hospital of Psychiatry, Bolligenstrasse 111, Bern 60, 3000, Switzerland
| | - Tim Vanbellingen
- Neurocenter, Luzerner Kantonsspital, Spitalstrasse 31, Luzern 16, 6000, Switzerland.,Gerontechnology and Rehabilitation Group, University of Bern, Murtenstrasse 50, Bern, 3008, Switzerland
| | - Sebastian Walther
- University Hospital of Psychiatry, Bolligenstrasse 111, Bern 60, 3000, Switzerland
| | - Stephan Bohlhalter
- Neurocenter, Luzerner Kantonsspital, Spitalstrasse 31, Luzern 16, 6000, Switzerland.,Department of Clinical Research, University of Bern, Bern, 3000, Switzerland
| |
Collapse
|
38
|
Mubeen AM, Ardekani B, Tagliati M, Alterman R, Dhawan V, Eidelberg D, Sidtis JJ. Global and multi-focal changes in cerebral blood flow during subthalamic nucleus stimulation in Parkinson's disease. J Cereb Blood Flow Metab 2018; 38:697-705. [PMID: 28421851 PMCID: PMC5888853 DOI: 10.1177/0271678x17705042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Electrical stimulation of subthalamic nuclei (STN) is a widely used therapy in Parkinson's disease (PD). While deep brain stimulation (DBS) of the STN alters the neurophysiological activity in basal ganglia, the therapeutic mechanism has not been established. A positron emission tomography (PET) study of cerebral blood flow (CBF) during speech production in PD subjects treated with STN-DBS found significant increases in global (whole-brain) CBF.1 That study utilized a series of whole-slice regions of interest to obtain global CBF values. The present study examined this effect using a voxel-based principal component analysis (PCA) combined with Fisher's linear discriminant analysis (FLDA) to classify STN-DBS on versus STN-DBS off whole-brain images. The approach yielded wide-spread CBF changes that classified STN-DBS status with accuracy, sensitivity, and specificity approaching 90%. The PCA component of the analysis supported the observation of a global CBF change during STN-DBS. The FLDA component demonstrated wide-spread multi-focal CBF changes. Further, CBF measurements related to a number of subject characteristics when STN-DBS was off, but not when it was on, suggesting that the normal relationship between CBF and behavior may be disrupted by this form of neuromodulation.
Collapse
Affiliation(s)
- Asim M Mubeen
- 1 Brain and Behavior Laboratory, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Babak Ardekani
- 2 Center for Brain Imaging and Neuromodulation, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Michele Tagliati
- 3 Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Alterman
- 4 Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - John J Sidtis
- 1 Brain and Behavior Laboratory, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
39
|
Modarres HP, Janmaleki M, Novin M, Saliba J, El-Hajj F, RezayatiCharan M, Seyfoori A, Sadabadi H, Vandal M, Nguyen MD, Hasan A, Sanati-Nezhad A. In vitro models and systems for evaluating the dynamics of drug delivery to the healthy and diseased brain. J Control Release 2018; 273:108-130. [PMID: 29378233 DOI: 10.1016/j.jconrel.2018.01.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) plays a crucial role in maintaining brain homeostasis and transport of drugs to the brain. The conventional animal and Transwell BBB models along with emerging microfluidic-based BBB-on-chip systems have provided fundamental functionalities of the BBB and facilitated the testing of drug delivery to the brain tissue. However, developing biomimetic and predictive BBB models capable of reasonably mimicking essential characteristics of the BBB functions is still a challenge. In addition, detailed analysis of the dynamics of drug delivery to the healthy or diseased brain requires not only biomimetic BBB tissue models but also new systems capable of monitoring the BBB microenvironment and dynamics of barrier function and delivery mechanisms. This review provides a comprehensive overview of recent advances in microengineering of BBB models with different functional complexity and mimicking capability of healthy and diseased states. It also discusses new technologies that can make the next generation of biomimetic human BBBs containing integrated biosensors for real-time monitoring the tissue microenvironment and barrier function and correlating it with the dynamics of drug delivery. Such integrated system addresses important brain drug delivery questions related to the treatment of brain diseases. We further discuss how the combination of in vitro BBB systems, computational models and nanotechnology supports for characterization of the dynamics of drug delivery to the brain.
Collapse
Affiliation(s)
- Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Mohsen Janmaleki
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Mana Novin
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - John Saliba
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Fatima El-Hajj
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Mahdi RezayatiCharan
- Breast Cancer Research Center (BCRC), ACECR, Tehran, Iran; School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Amir Seyfoori
- Breast Cancer Research Center (BCRC), ACECR, Tehran, Iran; School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamid Sadabadi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Milène Vandal
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Anwarul Hasan
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, 2713, Qatar
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada.
| |
Collapse
|
40
|
Lerner RP, Francardo V, Fujita K, Bimpisidis Z, Jourdain VA, Tang CC, Dewey SL, Chaly T, Cenci MA, Eidelberg D. Levodopa-induced abnormal involuntary movements correlate with altered permeability of the blood-brain-barrier in the basal ganglia. Sci Rep 2017; 7:16005. [PMID: 29167476 PMCID: PMC5700135 DOI: 10.1038/s41598-017-16228-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/09/2017] [Indexed: 01/24/2023] Open
Abstract
Chronic levodopa treatment leads to the appearance of dyskinesia in the majority of Parkinson’s disease patients. Neurovascular dysregulation in putaminal and pallidal regions is thought to be an underlying feature of this complication of treatment. We used microPET to study unilaterally lesioned 6-hydroxydopamine rats that developed levodopa-induced abnormal involuntary movements (AIMs) after three weeks of drug treatment. Animals were scanned with [15O]-labeled water and [18F]-fluorodeoxyglucose, to map regional cerebral blood flow and glucose metabolism, and with [11C]-isoaminobutyric acid (AIB), to assess blood-brain-barrier (BBB) permeability, following separate injections of levodopa or saline. Multitracer scan data were acquired in each animal before initiating levodopa treatment, and again following the period of daily drug administration. Significant dissociation of vasomotor and metabolic levodopa responses was seen in the striatum/globus pallidus (GP) of the lesioned hemisphere. These changes were accompanied by nearby increases in [11C]-AIB uptake in the ipsilateral GP, which correlated with AIMs scores. Histopathological analysis revealed high levels of microvascular nestin immunoreactivity in the same region. The findings demonstrate that regional flow-metabolism dissociation and increased BBB permeability are simultaneously induced by levodopa within areas of active microvascular remodeling, and that such changes correlate with the severity of dyskinesia.
Collapse
Affiliation(s)
- Renata P Lerner
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Koji Fujita
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Zisis Bimpisidis
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Vincent A Jourdain
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Chris C Tang
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Stephen L Dewey
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Thomas Chaly
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
| |
Collapse
|
41
|
Jourdain VA, Schindlbeck KA, Tang CC, Niethammer M, Choi YY, Markowitz D, Nazem A, Nardi D, Carras N, Feigin A, Ma Y, Peng S, Dhawan V, Eidelberg D. Increased putamen hypercapnic vasoreactivity in levodopa-induced dyskinesia. JCI Insight 2017; 2:96411. [PMID: 29046477 DOI: 10.1172/jci.insight.96411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/07/2017] [Indexed: 12/29/2022] Open
Abstract
In a rodent model of Parkinson's disease (PD), levodopa-induced involuntary movements have been linked to striatal angiogenesis - a process that is difficult to document in living human subjects. Angiogenesis can be accompanied by localized increases in cerebral blood flow (CBF) responses to hypercapnia. We therefore explored the possibility that, in the absence of levodopa, local hypercapnic CBF responses are abnormally increased in PD patients with levodopa-induced dyskinesias (LID) but not in their nondyskinetic (NLID) counterparts. We used H215O PET to scan 24 unmedicated PD subjects (12 LID and 12 NLID) and 12 matched healthy subjects in the rest state under normocapnic and hypercapnic conditions. Hypercapnic CBF responses were compared to corresponding levodopa responses from the same subjects. Group differences in hypercapnic vasoreactivity were significant only in the posterior putamen, with greater CBF responses in LID subjects compared with the other subjects. Hypercapnic and levodopa-mediated CBF responses measured in this region exhibited distinct associations with disease severity: the former correlated with off-state motor disability ratings but not symptom duration, whereas the latter correlated with symptom duration but not motor disability. These are the first in vivo human findings linking LID to microvascular changes in the basal ganglia.
Collapse
Affiliation(s)
- Vincent A Jourdain
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Katharina A Schindlbeck
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Chris C Tang
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Martin Niethammer
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| | - Yoon Young Choi
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | | | - Amir Nazem
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Dominic Nardi
- Department of Anesthesiology, Northwell Health, Manhasset, New York, USA
| | - Nicholas Carras
- Department of Anesthesiology, Northwell Health, Manhasset, New York, USA
| | - Andrew Feigin
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| | - Yilong Ma
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Shichun Peng
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Vijay Dhawan
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - David Eidelberg
- Center for Neurosciences, Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Neurology, Northwell Health, Manhasset, New York, USA
| |
Collapse
|
42
|
Al-Bachari S, Vidyasagar R, Emsley HC, Parkes LM. Structural and physiological neurovascular changes in idiopathic Parkinson's disease and its clinical phenotypes. J Cereb Blood Flow Metab 2017; 37:3409-3421. [PMID: 28112022 PMCID: PMC5624390 DOI: 10.1177/0271678x16688919] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neurovascular changes are likely to interact importantly with the neurodegenerative process in idiopathic Parkinson's disease (IPD). Markers of neurovascular status (NVS) include white matter lesion (WML) burden and arterial spin labelling (ASL) measurements of cerebral blood flow (CBF) and arterial arrival time (AAT). We investigated NVS in IPD, including an analysis of IPD clinical phenotypes, by comparison with two control groups, one with a history of clinical cerebrovascular disease (CVD) (control positive, CP) and one without CVD (control negative, CN). Fifty-one patients with IPD (mean age 69.0 ± 7.7 years) (21 tremor dominant (TD), 24 postural instability and gait disorder (PIGD) and six intermediates), 18 CP (mean age 70.1 ± 8.0 years) and 34 CN subjects (mean age 67.4 ± 7.6 years) completed a 3T MRI scan protocol including T2-weighted fluid-attenuated inversion recovery (FLAIR) and ASL. IPD patients showed diffuse regions of significantly prolonged AAT, small regions of lower CBF and greater WML burden by comparison with CN subjects. TD patients showed lower WML volume by comparison with PIGD patients. These imaging data thus show altered NVS in IPD, with some evidence for IPD phenotype-specific differences.
Collapse
Affiliation(s)
- Sarah Al-Bachari
- 1 Department of Neurology, Salford Royal NHS Foundation Trust, Salford, UK.,2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,3 Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Rishma Vidyasagar
- 2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,4 Anatomy and Neuroscience Department, University of Melbourne, Melbourne, Australia.,5 Florey Institute of Neuroscience and Mental Health, Heidelberg, Melbourne, Australia
| | - Hedley Ca Emsley
- 6 Department of Neurology, Royal Preston Hospital, Preston, UK.,7 Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Laura M Parkes
- 2 Division of Informatics, Imaging and Data Sciences, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,8 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
Bimpisidis Z, Öberg CM, Maslava N, Cenci MA, Lundblad C. Differential effects of gaseous versus injectable anesthetics on changes in regional cerebral blood flow and metabolism induced by l-DOPA in a rat model of Parkinson's disease. Exp Neurol 2017; 292:113-124. [PMID: 28284817 DOI: 10.1016/j.expneurol.2017.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 01/03/2023]
Abstract
Preclinical imaging of brain activity requires the use of anesthesia. In this study, we have compared the effects of two widely used anesthetics, inhaled isoflurane and ketamine/xylazine cocktail, on cerebral blood flow and metabolism in a rat model of Parkinson's disease and l-DOPA-induced dyskinesia. Specific tracers were used to estimate regional cerebral blood flow (rCBF - [14C]-iodoantipyrine) and regional cerebral metabolic rate (rCMR - [14C]-2-deoxyglucose) with a highly sensitive autoradiographic method. The two types of anesthetics had quite distinct effects on l-DOPA-induced changes in rCBF and rCMR. Isoflurane did not affect either the absolute rCBF values or the increases in rCBF in the basal ganglia after l-DOPA administration. On the contrary, rats anesthetized with ketamine/xylazine showed lower absolute rCBF values, and the rCBF increases induced by l-DOPA were masked. We developed a novel improved model to calculate rCMR, and found lower metabolic activities in rats anesthetized with isoflurane compared to animals anesthetized with ketamine/xylazine. Both anesthetics prevented changes in rCMR upon l-DOPA administration. Pharmacological challenges in isoflurane-anesthetized rats indicated that drugs mimicking the actions of ketamine/xylazine on adrenergic or glutamate receptors reproduced distinct effects of the injectable anesthetics on rCBF and rCMR. Our results highlight the importance of anesthesia in studies of cerebral flow and metabolism, and provide novel insights into mechanisms mediating abnormal neurovascular responses to l-DOPA in Parkinson's disease.
Collapse
Affiliation(s)
- Zisis Bimpisidis
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Carl M Öberg
- Department of Clinical Sciences, Nephrology, Lund University, Lund, Sweden
| | - Natallia Maslava
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Cornelia Lundblad
- Anesthesiology and Intensive Care, Department of Clinical Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Del-Bel E, Bortolanza M, Dos-Santos-Pereira M, Bariotto K, Raisman-Vozari R. l-DOPA-induced dyskinesia in Parkinson's disease: Are neuroinflammation and astrocytes key elements? Synapse 2016; 70:479-500. [DOI: 10.1002/syn.21941] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Elaine Del-Bel
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Mariza Bortolanza
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Maurício Dos-Santos-Pereira
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
| | - Keila Bariotto
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Rita Raisman-Vozari
- INSERM UMR 1127, CNRS UMR 7225, UPMC; Thérapeutique Expérimentale de la Neurodégénérescence, Hôpital de la Salpetrière-ICM (Institut du cerveau et de la moelle épinière); Paris France
| |
Collapse
|
45
|
Jourdain VA, Tang CC, Holtbernd F, Dresel C, Choi YY, Ma Y, Dhawan V, Eidelberg D. Flow-metabolism dissociation in the pathogenesis of levodopa-induced dyskinesia. JCI Insight 2016; 1:e86615. [PMID: 27699242 DOI: 10.1172/jci.insight.86615] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Levodopa-induced dyskinesia (LID) is the most common, disruptive complication of Parkinson's disease (PD) pharmacotherapy, yet despite decades of research, the changes in regional brain function underlying LID remain largely unknown. We previously found that the cerebral vasomotor and metabolic responses to levodopa are dissociated in PD subjects. Nonetheless, it is unclear whether levodopa-mediated dissociation is exaggerated in LID or distinguishes LID from non-LID subjects. To explore this possibility, we used dual-tracer positron emission tomography to quantify regional cerebral blood flow and metabolic activity in 28 PD subjects (14 LID, 14 non-LID), scanned before and during intravenous levodopa infusion. Levodopa-mediated dissociation was most prominent in the posterior putamen (P < 0.0001) and greater in LID than in non-LID and test-retest subjects. Strikingly, LID subjects also showed increased sensorimotor cortex (SMC) activity in the baseline, unmedicated state. Imaging data from an independent PD sample (106 subjects) linked these differences to loss of mesocortical dopamine terminals in advanced patients. In aggregate, the data suggest that LID results from an overactive vasomotor response to levodopa in the putamen on a background of disease-related increases in SMC activity. LID may thus be amenable to treatment that modulates the function of these 2 regions.
Collapse
|
46
|
Lerner RP, Bimpisidis Z, Agorastos S, Scherrer S, Dewey SL, Cenci MA, Eidelberg D. Dissociation of metabolic and hemodynamic levodopa responses in the 6-hydroxydopamine rat model. Neurobiol Dis 2016; 96:31-37. [PMID: 27544483 DOI: 10.1016/j.nbd.2016.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/03/2016] [Accepted: 08/16/2016] [Indexed: 11/17/2022] Open
Abstract
Dissociation of vasomotor and metabolic responses to levodopa has been observed in human subjects with Parkinson's disease (PD) studied with PET and in autoradiograms from 6-hydroxydopamine (6-OHDA) rat. In both species, acute levodopa administration was associated with increases in basal ganglia cerebral blood flow (CBF) with concurrent reductions in cerebral metabolic rate (CMR) for glucose in the same brain regions. In this study, we used a novel dual-tracer microPET technique to measure CBF and CMR levodopa responses in the same animal. Rats with unilateral 6-OHDA or sham lesion underwent sequential 15O-water (H215O) and 18F-fluorodeoxyglucose (FDG) microPET to map CBF and CMR following the injection of levodopa or saline. A subset of animals was separately scanned under ketamine/xylazine and isoflurane to compare the effects of these anesthetics. Regardless of anesthetic agent, 6-OHDA animals exhibited significant dissociation of vasomotor (ΔCBF) and metabolic (ΔCMR) responses to levodopa, with stereotyped increases in CBF and reductions in CMR in the basal ganglia ipsilateral to the dopamine lesion. No significant changes were seen in sham-lesioned animals. These data faithfully recapitulate analogous dissociation effects observed previously in human PD subjects scanned sequentially during levodopa infusion. This approach may have utility in the assessment of new drugs targeting the exaggerated regional vasomotor responses seen in human PD and in experimental models of levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Renata P Lerner
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
| | - Zisis Bimpisidis
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Stergiani Agorastos
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
| | - Sandra Scherrer
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
| | - Stephen L Dewey
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA.
| |
Collapse
|
47
|
Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L. Nanoparticle-mediated brain drug delivery: Overcoming blood–brain barrier to treat neurodegenerative diseases. J Control Release 2016; 235:34-47. [DOI: 10.1016/j.jconrel.2016.05.044] [Citation(s) in RCA: 917] [Impact Index Per Article: 101.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
|
48
|
Vo A, Sako W, Fujita K, Peng S, Mattis PJ, Skidmore FM, Ma Y, Uluğ AM, Eidelberg D. Parkinson's disease-related network topographies characterized with resting state functional MRI. Hum Brain Mapp 2016; 38:617-630. [PMID: 27207613 DOI: 10.1002/hbm.23260] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/24/2016] [Accepted: 05/03/2016] [Indexed: 11/10/2022] Open
Abstract
Spatial covariance mapping can be used to identify and measure the activity of disease-related functional brain networks. While this approach has been widely used in the analysis of cerebral blood flow and metabolic PET scans, it is not clear whether it can be reliably applied to resting state functional MRI (rs-fMRI) data. In this study, we present a novel method based on independent component analysis (ICA) to characterize specific network topographies associated with Parkinson's disease (PD). Using rs-fMRI data from PD and healthy subjects, we used ICA with bootstrap resampling to identify a PD-related pattern that reliably discriminated the two groups. This topography, termed rs-MRI PD-related pattern (fPDRP), was similar to previously characterized disease-related patterns identified using metabolic PET imaging. Following pattern identification, we validated the fPDRP by computing its expression in rs-fMRI testing data on a prospective case basis. Indeed, significant increases in fPDRP expression were found in separate sets of PD and control subjects. In addition to providing a similar degree of group separation as PET, fPDRP values correlated with motor disability and declined toward normal with levodopa administration. Finally, we used this approach in conjunction with neuropsychological performance measures to identify a separate PD cognition-related pattern in the patients. This pattern, termed rs-fMRI PD cognition-related pattern (fPDCP), was topographically similar to its PET-derived counterpart. Subject scores for the fPDCP correlated with executive function in both training and testing data. These findings suggest that ICA can be used in conjunction with bootstrap resampling to identify and validate stable disease-related network topographies in rs-fMRI. Hum Brain Mapp 38:617-630, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- An Vo
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Wataru Sako
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Koji Fujita
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Shichun Peng
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Paul J Mattis
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York.,Department of Neurology, Northwell Health, Manhasset, New York
| | - Frank M Skidmore
- Department of Neurology, University of Alabama School of Medicine, Birmingham, Alabama
| | - Yilong Ma
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Aziz M Uluğ
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institute for Medical Research, Manhasset, New York
| |
Collapse
|
49
|
Thiollier T, Wu C, Contamin H, Li Q, Zhang J, Bezard E. Permeability of blood-brain barrier in macaque model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson disease. Synapse 2016; 70:231-9. [PMID: 26799359 DOI: 10.1002/syn.21889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/06/2015] [Accepted: 01/18/2016] [Indexed: 01/16/2023]
Abstract
Brain bioavailability of drugs developed to address central nervous system diseases is classically documented through cerebrospinal fluid collected in normal animals, i.e., through an approximation as there are fundamental differences between cerebrospinal fluid and tissue contents. The fact that disease might affect brain availability of drugs is almost never considered at this stage although several conditions are associated with blood-brain barrier damage. Building upon our expertise in Parkinson's disease translational research, the present study addressed this gap comparing plasma and cerebrospinal fluid bioavailability of l-3,4-dihydroxyphenylalanine, carbamazepine, quinidine, lovastatin, and simvastatin, in healthy and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated macaques, the gold standard model of Parkinson's disease. The drugs were selected based upon their differential transport across the blood-brain barrier. Interestingly, brain bioavailability of quinidine was decreased while others were unaffected. Pharmacokinetics and pharmacodynamics experiments of drugs addressing Parkinson's disease might thus be performed in healthy animals unless the drugs are known to interact with the organic cation transporter.
Collapse
Affiliation(s)
- Thibaud Thiollier
- Cynbiose, Marcy l'Etoile, France.,Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000, Bordeaux, France
| | - Caisheng Wu
- Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China, 100050
| | | | - Qin Li
- Motac Neuroscience, Manchester, United Kingdom.,Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jinlan Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, People's Republic of China, 100050
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000, Bordeaux, France.,Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
50
|
Siddiqi SH, Abraham NK, Geiger CL, Karimi M, Perlmutter JS, Black KJ. The Human Experience with Intravenous Levodopa. Front Pharmacol 2016; 6:307. [PMID: 26779024 PMCID: PMC4701937 DOI: 10.3389/fphar.2015.00307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 12/11/2015] [Indexed: 11/24/2022] Open
Abstract
Objective: To compile a comprehensive summary of published human experience with levodopa given intravenously, with a focus on information required by regulatory agencies. Background: While safe intravenous (IV) use of levodopa has been documented for over 50 years, regulatory supervision for pharmaceuticals given by a route other than that approved by the U.S. Food and Drug Administration (FDA) has become increasingly cautious. If delivering a drug by an alternate route raises the risk of adverse events, an investigational new drug (IND) application is required, including a comprehensive review of toxicity data. Methods: Over 200 articles referring to IV levodopa were examined for details of administration, pharmacokinetics, benefit, and side effects. Results: We identified 142 original reports describing IVLD use in humans, beginning with psychiatric research in 1959–1960 before the development of peripheral decarboxylase inhibitors. At least 2760 subjects have received IV levodopa, and reported outcomes include parkinsonian signs, sleep variables, hormone levels, hemodynamics, CSF amino acid composition, regional cerebral blood flow, cognition, perception and complex behavior. Mean pharmacokinetic variables were summarized for 49 healthy subjects and 190 with Parkinson's disease. Side effects were those expected from clinical experience with oral levodopa and dopamine agonists. No articles reported deaths or induction of psychosis. Conclusion: At least 2760 patients have received IV levodopa with a safety profile comparable to that seen with oral administration.
Collapse
Affiliation(s)
- Shan H Siddiqi
- Department of Psychiatry, Washington University School of Medicine St. Louis, MO, USA
| | - Natalia K Abraham
- School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa Ottawa, ON, Canada
| | | | - Morvarid Karimi
- Department of Neurology, Washington University School of Medicine St. Louis, MO, USA
| | - Joel S Perlmutter
- Programs in Occupational Therapy and Physical Therapy, Division of Biology and Biomedical Sciences, Departments of Neurology, Radiology, and Anatomy and Neurobiology, Washington University School of Medicine St. Louis, MO, USA
| | - Kevin J Black
- Division of Biology and Biomedical Sciences, Departments of Psychiatry, Neurology, Radiology, and Anatomy and Neurobiology, Washington University School of Medicine St. Louis, MO, USA
| |
Collapse
|