101
|
Jung IH, Chang KW, Park SH, Chang WS, Jung HH, Chang JW. Complications After Deep Brain Stimulation: A 21-Year Experience in 426 Patients. Front Aging Neurosci 2022; 14:819730. [PMID: 35462695 PMCID: PMC9022472 DOI: 10.3389/fnagi.2022.819730] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDeep brain stimulation is an established treatment for movement disorders such as Parkinson’s disease, essential tremor, and dystonia. However, various complications that occur after deep brain stimulation are a major concern for patients and neurosurgeons.ObjectiveThis study aimed to analyze various complications that occur after deep brain stimulation.MethodsWe reviewed the medical records of patients with a movement disorder who underwent bilateral deep brain stimulation between 2000 and 2020. Among them, patients requiring revision surgery were analyzed.ResultsA total of 426 patients underwent bilateral deep brain stimulation for a movement disorder. The primary disease was Parkinson’s disease in 315 patients, followed by dystonia in 71 patients and essential tremor in 40 patients. Twenty-six (6.1%) patients had complications requiring revision surgery; the most common complication was infection (12 patients, 2.8%).ConclusionVarious complications may occur after deep brain stimulation, and patient prognosis should be improved by reducing complications.
Collapse
Affiliation(s)
- In-Ho Jung
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurosurgery, Dankook University College of Medicine, Cheonan, South Korea
| | - Kyung Won Chang
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - So Hee Park
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Won Seok Chang
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Ho Jung
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Chang
- Department of Neurosurgery, Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Jin Woo Chang,
| |
Collapse
|
102
|
Faraji B, Gheisarnejad M, Esfahani Z, Khooban MH. Smart Sensor Control for Rehabilitation in Parkinson's Patients. IEEE TRANSACTIONS ON EMERGING TOPICS IN COMPUTATIONAL INTELLIGENCE 2022. [DOI: 10.1109/tetci.2020.3045483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
103
|
Shah J, Quinkert C, Collar B, Williams M, Biggs E, Irazoqui P. A Highly Miniaturized, Chronically Implanted ASIC for Electrical Nerve Stimulation. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2022; 16:233-243. [PMID: 35201991 PMCID: PMC9195150 DOI: 10.1109/tbcas.2022.3153282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
We present a wireless, fully implantable device for electrical stimulation of peripheral nerves consisting of a powering coil, a tuning network, a Zener diode, selectable stimulation parameters, and a stimulator IC, all encapsulated in biocompatible silicone. A wireless RF signal at 13.56 MHz powers the implant through the on-chip rectifier. The ASIC, designed in TSMC's 180 nm MS RF G process, occupies an area of less than 1.2 mm2. The IC enables externally selectable current-controlled stimulation through an on-chip read-only memory with a wide range of 32 stimulation parameters (90-750 µA amplitude, 100 µs or 1 ms pulse width, 15 or 50 Hz frequency). The IC generates the constant current waveform using an 8-bit binary weighted DAC and an H-Bridge. At the most power-hungry stimulation parameter, the average power consumption during a stimulus pulse is 2.6 mW with a power transfer efficiency of ∼5.2%. In addition to benchtop and acute testing, we chronically implanted two versions of the device (a design with leads and a leadless design) on two rats' sciatic nerves to verify the long-term efficacy of the IC and the full system. The leadless device had the following dimensions: height of 0.45 cm, major axis of 1.85 cm, and minor axis of 1.34 cm, with similar dimensions for the device with leads. Both devices were implanted and worked for experiments lasting from 21-90 days. To the best of our knowledge, the fabricated IC is the smallest constant-current stimulator that has been tested chronically.
Collapse
|
104
|
Cole RC, Okine DN, Yeager BE, Narayanan NS. Neuromodulation of cognition in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:435-455. [PMID: 35248205 DOI: 10.1016/bs.pbr.2022.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Neuromodulation is a widely used treatment for motor symptoms of Parkinson's disease (PD). It can be a highly effective treatment as a result of knowledge of circuit dysfunction associated with motor symptoms in PD. However, the mechanisms underlying cognitive symptoms of PD are less well-known, and the effects of neuromodulation on these symptoms are less consistent. Nonetheless, neuromodulation provides a unique opportunity to modulate motor and cognitive circuits while minimizing off-target side effects. We review the modalities of neuromodulation used in PD and the potential implications for cognitive symptoms. There have been some encouraging findings with both invasive and noninvasive modalities of neuromodulation, and there are promising advances being made in the field of therapeutic neuromodulation. Substantial work is needed to determine which modulation targets are most effective for the different types of cognitive deficits of PD.
Collapse
Affiliation(s)
- Rachel C Cole
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | - Derrick N Okine
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | - Brooke E Yeager
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | | |
Collapse
|
105
|
Wu YX, Xiang W, Wang JJ, Liu XM, Yi DY, Tian H, Zhao HY, Jiang XB, Fu P. A Modified Dura Puncture Procedure to Reduce Brain Shift in Deep Brain Stimulation Surgery: One Institution's Experience. Front Neurol 2022; 13:845926. [PMID: 35295828 PMCID: PMC8920348 DOI: 10.3389/fneur.2022.845926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
Objective The therapeutic effect of deep brain stimulation (DBS) surgery mainly depends on the accuracy of electrode placement and the reduction in brain shift. Among the standard procedures, cerebrospinal fluid (CSF) loss or pneumocephalus caused by dura incision (DI) is thought to be the main reason for brain shift and inaccuracy of electrode placement. In the current study, we described a modified dura puncture (DP) procedure to reduce brain shift and compare it with the general procedure of DBS surgery in terms of electrode placement accuracy. Materials and Methods We retrospectively analyzed a series of 132 patients who underwent DBS surgery in Wuhan Union Hospital from December 2015 to April 2021. According to the different surgery procedures, patients were classified into two cohorts: the DI group (DI cohort) had 49 patients who receive the general procedure, and the DP group (DP cohort) had 83 patients who receive the modified procedure. Postoperative pneumocephalus volume (PPV) and CSF loss volume, electrode fusion error (EFE), and trajectory number were calculated. Meanwhile, intraoperative electrophysiological signal length (IESL), electrode implantation duration, and other parameters were analyzed. Results In the current study, we introduced an improved electrode implantation procedure for DBS surgery named the DP procedure. Compared with the general DI cohort (n = 49), the modified DP cohort (n = 83) had a shorter electrode implantation duration (p < 0.0001), smaller PPV, lower CSF leakage volume (p < 0.0001), and smaller EFE (p < 0.0001). There was no significant difference in IESL (p > 0.05) or adverse events (perioperative cerebral haematoma, skin erosion, epilepsy, p > 0.05) between the two cohorts. Conclusion The DP procedure is a modified procedure that can reduce brain shift and ensure implantation accuracy during DBS surgery without adverse events.
Collapse
Affiliation(s)
- Yu-Xi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Jing Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Ming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong-Ye Yi
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Tian
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Yang Zhao
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Bing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Peng Fu
| |
Collapse
|
106
|
de Noordhout AM, Mouchamps M, Remacle JM, Delstanche S, Bonhomme V, Gonce M. Subthalamic deep brain stimulation versus best medical treatment: a 12-year follow-up. Acta Neurol Belg 2022; 122:197-202. [PMID: 35084704 PMCID: PMC8894213 DOI: 10.1007/s13760-022-01874-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/17/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Electrical stimulation of the sub-thalamic nucleus (STN-DBS) is well established to alleviate motor fluctuations in advanced Parkinson's disease but little is known about its very long-term efficacy. METHODS We followed over 12 years 15 parkinsonian patients having undergone STN-DBS and compared them to a matched group of 14 patients with best medical drug therapy. All had been considered as good candidates for surgery. They were allocated to each group depending on their own decision. RESULTS After 12 years, mortality rates were similar in both groups. In the DBS group, best "on" UPDRS III scores (on medications, on stimulation) remained significantly better and dyskinesia shorter and weaker than in the drug-treated group (on medication only). Yet, looking at independent life and quality of life (QoL) evaluated with PDQ39, no significant difference could be observed between groups at the end of follow-up, probably due to development of dopa- and stimulation-resistant motor and non-motor symptoms like falls, freezing, dementia, apathy and depression, the latter two more frequent in the DBS group. CONCLUSION Drug- and DBS-resistant symptoms and signs occur more often after long disease evolution and in elder patients. It might be why differences in QoL between both groups no longer existed after twelve years as, compared to other studies, our patients were older at inclusion.
Collapse
Affiliation(s)
| | | | | | - Stéphanie Delstanche
- University Department of Neurology, Hôpital de la Citadelle, 4000, Liège, Belgium
| | - Vincent Bonhomme
- Department of Anaesthesiology, Hôpital de la Citadelle, 4000, Liège, Belgium
| | - Michel Gonce
- University Department of Neurology, Hôpital de la Citadelle, 4000, Liège, Belgium
| |
Collapse
|
107
|
AIM in Neurodegenerative Diseases: Parkinson and Alzheimer. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
108
|
Oh T, Daadi ES, Kim J, Daadi EW, Chen PJ, Roy-Choudhury G, Bohmann J, Blass BE, Daadi MM. Dopamine D3 receptor ligand suppresses the expression of levodopa-induced dyskinesia in nonhuman primate model of parkinson's disease. Exp Neurol 2022; 347:113920. [PMID: 34762921 DOI: 10.1016/j.expneurol.2021.113920] [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] [Received: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/04/2022]
Abstract
Parkinson's disease (PD) is a complex multisystem, chronic and so far incurable disease with significant unmet medical needs. The incidence of PD increases with aging and the expected burden will continue to escalate with our aging population. Since its discovery in the 1961 levodopa has remained the gold standard pharmacotherapy for PD. However, the progressive nature of the neurodegenerative process in and beyond the nigrostriatal system causes a multitude of side effects, including levodopa-induced dyskinesia within 5 years of therapy. Attenuating dyskinesia has been a significant challenge in the clinical management of PD. We report on a small molecule that eliminates the expression of levodopa-induced dyskinesia and significantly improves PD-like symptoms. The lead compound PD13R we discovered is a dopamine D3 receptor partial agonist with high affinity and selectivity, orally active and with desirable drug-like properties. Future studies are aimed at developing this lead compound for treating PD patients with dyskinesia.
Collapse
Affiliation(s)
- Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Elyas S Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA; Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Etienne W Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Peng-Jen Chen
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Gourav Roy-Choudhury
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Benjamin E Blass
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Marcel M Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA; Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA; Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
109
|
Steinmüller JB, Bjarkam CR, Orlowski D, Sørensen JCH, Glud AN. Anterograde Tracing From the Göttingen Minipig Motor and Prefrontal Cortex Displays a Topographic Subthalamic and Striatal Axonal Termination Pattern Comparable to Previous Findings in Primates. Front Neural Circuits 2021; 15:716145. [PMID: 34899195 PMCID: PMC8661455 DOI: 10.3389/fncir.2021.716145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/02/2021] [Indexed: 01/20/2023] Open
Abstract
Background: Deep brain stimulation (DBS) of the dorsal subthalamic nucleus (STN) is a validated neurosurgical treatment of Parkinson’s Disease (PD). To investigate the mechanism of action, including potential DBS induced neuroplasticity, we have previously used a minipig model of Parkinson’s Disease, although the basal ganglia circuitry was not elucidated in detail. Aim: To describe the cortical projections from the primary motor cortex (M1) to the basal ganglia and confirm the presence of a cortico-striatal pathway and a hyperdirect pathway to the subthalamic nucleus, respectively, which is known to exist in primates. Materials and Methods: Five female Göttingen minipigs were injected into the primary motor cortex (n = 4) and adjacent prefrontal cortex (n = 1) with the anterograde neuronal tracer, Biotinylated Dextran Amine (BDA). 4 weeks later the animals were sacrificed and the brains cryosectioned into 30 μm thick coronal sections for subsequent microscopic analysis. Results: The hyperdirect axonal connections from the primary motor cortex were seen to terminate in the dorsolateral STN, whereas the axonal projections from the prefrontal cortex terminated medially in the STN. Furthermore, striatal tracing from the motor cortex was especially prominent in the dorsolateral putamen and less so in the dorsolateral caudate nucleus. The prefrontal efferents were concentrated mainly in the caudate nucleus and to a smaller degree in the juxtacapsular dorsal putamen, but they were also found in the nucleus accumbens and ventral prefrontal cortex. Discussion: The organization of the Göttingen minipig basal ganglia circuitry is in accordance with previous descriptions in primates. The existence of a cortico-striatal and hyperdirect basal ganglia pathway in this non-primate, large animal model may accordingly permit further translational studies on STN-DBS induced neuroplasticity of major relevance for future DBS treatments.
Collapse
Affiliation(s)
- Johannes Bech Steinmüller
- CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | | | - Dariusz Orlowski
- CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Jens Christian Hedemann Sørensen
- CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Andreas Nørgaard Glud
- CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
110
|
Sadighi M, Şişman M, Eyüboğlu BM. SNR and total acquisition time analysis of multi-echo FLASH pulse sequence for current density imaging. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2021; 333:107098. [PMID: 34794090 DOI: 10.1016/j.jmr.2021.107098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
Magnetic Resonance Current Density Imaging (MRCDI) is an imaging modality providing cross-sectional current density (J¯) information inside the body. The clinical applicability of MRCDI is highly dependent on the sensitivity of the acquired noisy current-induced magnetic flux density (B∼z) distributions. Here, a novel analysis is developed to investigate the combined effect of relevant parameters of the RF spoiled gradient echo (FLASH) pulse sequence on the SNR level and the total acquisition time (TAT) of the acquired B∼z images. The proposed analysis then is expanded for a multi-echo FLASH (ME-FLASH) pulse sequence to take advantage of combining the multiple echoes to achieve B∼zcomb distribution with a higher SNR than the one achievable with a single echo acquisition. The optimized sequence parameters to acquire a B∼z distribution with the highest possible SNR for a given acquisition time or the desired SNR in the shortest scan time are estimated using the proposed analysis. The analysis also provides different sets of sequence parameters to acquire B∼z distributions with the same SNR at almost the same TAT. Furthermore, the effects of intensive utilization of the gradients and the magnetohydrodynamic (MHD) flow velocity on the acquired B∼z distribution in MRCDI experiments is investigated. The analytical results of the proposed analysis are validated experimentally using an imaging phantom having the conductivity and the relaxation parameters of the brain white matter tissue.
Collapse
Affiliation(s)
- Mehdi Sadighi
- Department of Electrical and Electronics Engineering, Middle East Technical University, 06800 Ankara, Turkey.
| | - Mert Şişman
- Department of Electrical and Electronics Engineering, Middle East Technical University, 06800 Ankara, Turkey
| | - B Murat Eyüboğlu
- Department of Electrical and Electronics Engineering, Middle East Technical University, 06800 Ankara, Turkey
| |
Collapse
|
111
|
Tai CH. Subthalamic burst firing: A pathophysiological target in Parkinson's disease. Neurosci Biobehav Rev 2021; 132:410-419. [PMID: 34856222 DOI: 10.1016/j.neubiorev.2021.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/16/2021] [Accepted: 11/28/2021] [Indexed: 11/27/2022]
Abstract
Understanding the pathophysiological mechanism of Parkinson's disease (PD) in the subthalamic nucleus (STN) has become a critical issue since deep brain stimulation (DBS) in this region has been proven as an effective treatment for this disease. The STN possesses a special ability to switch from the spike to the burst firing mode in response to dopamine deficiency in parkinsonism, and this STN burst is considered an electrophysiological signature of the cortico-basal ganglia circuit in the brains of PD patients. This review focuses on the role of STN burst firing in the pathophysiology of PD and during DBS. Here, we review existing literature on how STN bursts originate and the specific factors affecting their formation; how STN burst firing causes motor symptoms in PD and how interventions can rescue these symptoms. Finally, the similarities and differences between the two electrophysiological hallmarks of PD, STN burst firing and beta-oscillation, are discussed. STN burst firing should be considered as a pathophysiological target in PD during treatment with DBS.
Collapse
Affiliation(s)
- Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, No. 7, Jhongshan South Road, 100225, Taipei, Taiwan.
| |
Collapse
|
112
|
Löser J, Luthardt J, Rullmann M, Weise D, Sabri O, Meixensberger J, Hesse S, Winkler D. Striatal dopamine transporter availability and individual clinical course within the 1-year follow-up of deep brain stimulation of the subthalamic nucleus in patients with Parkinson's disease. J Neurosurg 2021; 135:1429-1435. [PMID: 33607613 DOI: 10.3171/2020.8.jns192740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 08/07/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Degeneration of dopaminergic neurons in the substantia nigra projecting to the striatum is responsible for the motor symptoms in Parkinson's disease (PD). Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is a well-established procedure to alleviate these symptoms in advanced PD. Yet the mechanism of action, especially the effects of STN-DBS on the availability of striatal dopamine transporter (DAT) as a marker of nigrostriatal nerve cell function, remains largely unknown. The aim of this study was therefore to evaluate whether 1) DAT availability changes within 1 year of STN-DBS and 2) the clinical outcome can be predicted based on preoperative DAT availability. METHODS Twenty-seven PD patients (mean age 62.7 ± 8.9 years; mean duration of illness 13.0 ± 4.9 years; PD subtypes: akinetic-rigid, n = 11; equivalence, n = 13; and tremor-dominant, n = 3) underwent [123I]FP-CIT SPECT preoperatively and after 1 year of STN-DBS. DAT availability as determined by the specific binding ratio (SBR) was assessed by volume of interest (VOI) analysis of the caudate nucleus and the putamen ipsilateral and contralateral to the clinically more affected side. RESULTS Unified Parkinson's Disease Rating Scale (UPDRS) III scores improved significantly (mean preoperative on medication 25.6 ± 12.3, preoperative off medication 42.3 ± 15.2, postoperative on medication/off stimulation 41.4 ± 13.2, and postoperative on medication/on stimulation 16.1 ± 9.4; preoperative on medication vs postoperative on medication/on stimulation, p = 0.006), while the levodopa-equivalent daily dose was reduced (mean preoperative 957 ± 440 mg vs postoperative 313 ± 189 mg, p < 0.001). The SBR did not differ significantly before and 1 year after DBS, regardless of PD subtype. Preoperative DAT availability was not related to the change in UPDRS III score, but the change in DAT availability was significantly correlated with the change in UPDRS III score (contralateral head of the caudate VOI, p = 0.014; contralateral putamen VOI, p = 0.018). CONCLUSIONS Overall, DAT availability did not change significantly after 1 year of STN-DBS. However, on an individual basis, the improvement in UPDRS III score was associated with an increase in DAT availability, whereas DAT availability before STN-DBS surgery did not predict the clinical outcome. Whether a subtype-specific pattern of preoperative DAT availability can become a reliable predictor of successful STN-DBS must be evaluated in larger study cohorts.
Collapse
Affiliation(s)
- Julia Löser
- Departments of1Nuclear Medicine
- 2Neurosurgery, and
- 3Pediatric Surgery, University of Leipzig
| | | | - Michael Rullmann
- Departments of1Nuclear Medicine
- 4Integrated Treatment and Research Centre (IFB) Adiposity Diseases, University of Leipzig; and
| | - David Weise
- 5Department of Neurology, Asklepios Clinic Stadtroda, Germany
| | | | | | - Swen Hesse
- Departments of1Nuclear Medicine
- 4Integrated Treatment and Research Centre (IFB) Adiposity Diseases, University of Leipzig; and
| | | |
Collapse
|
113
|
Phokaewvarangkul O, Vateekul P, Wichakam I, Anan C, Bhidayasiri R. Using Machine Learning for Predicting the Best Outcomes With Electrical Muscle Stimulation for Tremors in Parkinson's Disease. Front Aging Neurosci 2021; 13:727654. [PMID: 34566628 PMCID: PMC8461308 DOI: 10.3389/fnagi.2021.727654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
Recent studies have identified that peripheral stimulation in Parkinson’s disease (PD) is effective in tremor reduction, indicating that a peripheral feedback loop plays an important role in the tremor reset mechanism. This was an open-label, quasi-experimental, pre- and post-test design, single-blind, single-group study involving 20 tremor-dominant PD patients. The objective of this study is to explore the effect of electrical muscle stimulation (EMS) as an adjunctive treatment for resting tremor during “on” period and to identify the best machine learning model to predict the suitable stimulation level that will yield the longest period of tremor reduction or tremor reset time. In this study, we used a Parkinson’s glove to evaluate, stimulate, and quantify the tremors of PD patients. This adjustable glove incorporates a 3-axis gyroscope to measure tremor signals and an EMS to provide an on-demand muscle stimulation to suppress tremors. Machine learning models were applied to identify the suitable pulse amplitude (stimulation level) in five classes that led to the longest tremor reset time. The study was registered at the www.clinicaltrials.gov under the name “The Study of Rest Tremor Suppression by Using Electrical Muscle Stimulation” (NCT02370108). Twenty tremor-dominant PD patients were recruited. After applying an average pulse amplitude of 6.25 (SD 2.84) mA and stimulation period of 440.7 (SD 560.82) seconds, the total time of tremor reduction, or tremor reset time, was 329.90 (SD 340.91) seconds. A significant reduction in tremor parameters during stimulation was demonstrated by a reduction of Unified Parkinson’s Disease Rating Scale (UPDRS) scores, and objectively, with a reduction of gyroscopic data (p < 0.05, each). None of the subjects reported any serious adverse events. We also compared gyroscopic data with five machine learning techniques: Logistic Regression, Random Forest, Support Vector Machine (SVM), Neural Network (NN), and Long-Short-Term-Memory (LSTM). The machine learning model that gave the highest accuracy was LSTM, which obtained: accuracy = 0.865 and macro-F1 = 0.736. This study confirms the efficacy of EMS in the reduction of resting tremors in PD. LSTM was identified as the most effective model for predicting pulse amplitude that would elicit the longest tremor reset time. Our study provides further insight on the tremor reset mechanism in PD.
Collapse
Affiliation(s)
- Onanong Phokaewvarangkul
- Department of Medicine, Faculty of Medicine, Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Peerapon Vateekul
- Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
| | - Itsara Wichakam
- Department of Medicine, Faculty of Medicine, Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chanawat Anan
- Department of Medicine, Faculty of Medicine, Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Roongroj Bhidayasiri
- Department of Medicine, Faculty of Medicine, Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand.,The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
114
|
Lachenmayer ML, Mürset M, Antih N, Debove I, Muellner J, Bompart M, Schlaeppi JA, Nowacki A, You H, Michelis JP, Dransart A, Pollo C, Deuschl G, Krack P. Subthalamic and pallidal deep brain stimulation for Parkinson's disease-meta-analysis of outcomes. NPJ PARKINSONS DISEASE 2021; 7:77. [PMID: 34489472 PMCID: PMC8421387 DOI: 10.1038/s41531-021-00223-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/12/2021] [Indexed: 12/26/2022]
Abstract
Although deep brain stimulation (DBS) of the globus pallidus internus (GPi) and the subthalamic nucleus (STN) has become an established treatment for Parkinson’s disease (PD), a recent meta-analysis of outcomes is lacking. To address this gap, we performed a meta-analysis of bilateral STN- and GPi-DBS studies published from 1990-08/2019. Studies with ≥10 subjects reporting Unified Parkinson’s Disease Rating Scale (UPDRS) III motor scores at baseline and 6–12 months follow-up were included. Several outcome variables were analyzed and adverse events (AE) were summarized. 39 STN studies (2035 subjects) and 5 GPi studies (292 subjects) were eligible. UPDRS-II score after surgery in the stimulation-ON/medication-OFF state compared to preoperative medication-OFF state improved by 47% with STN-DBS and 18.5% with GPi-DBS. UPDRS-III score improved by 50.5% with STN-DBS and 29.8% with GPi-DBS. STN-DBS improved dyskinesia by 64%, daily OFF time by 69.1%, and quality of life measured by PDQ-39 by 22.2%, while Levodopa Equivalent Daily Dose (LEDD) was reduced by 50.0%. For GPi-DBS information regarding dyskinesia, OFF time, PDQ-39 and LEDD was insufficient for further analysis. Correlation analysis showed that preoperative L-dopa responsiveness was highly predictive of the STN-DBS motor outcome across all studies. Most common surgery-related AE were infection (5.1%) and intracranial hemorrhage (3.1%). Despite a series of technological advances, outcomes of modern surgery are still comparable with those of the early days of DBS. Recent changes in target selection with a preference of GPi in elderly patients with cognitive deficits and more psychiatric comorbidities require more published data for validation.
Collapse
Affiliation(s)
- M Lenard Lachenmayer
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Melina Mürset
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Ines Debove
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Julia Muellner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Janine-Ai Schlaeppi
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andreas Nowacki
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hana You
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Joan P Michelis
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Claudio Pollo
- Department of Neurosurgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Guenther Deuschl
- Department of Neurology, UKSH, Christian-Albrechts-University, Kiel, Germany
| | - Paul Krack
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
115
|
Mei X, Ye D, Zhang F, Di C. Implantable application of polymer‐based biosensors. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xiangyuan Mei
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Dekai Ye
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| | - Fengjiao Zhang
- School of Chemical Sciences University of Chinese Academy of Sciences Beijing China
| | - Chong‐an Di
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids Institute of Chemistry, Chinese Academy of Sciences Beijing China
| |
Collapse
|
116
|
Radmard S, Ortega RA, Ford B, Vanegas-Arroyave N, McKhann GM, Sheth SA, Winfield L, Luciano MS, Saunders-Pullman R, Pullman SL. Using computerized spiral analysis to evaluate deep brain stimulation outcomes in Parkinson disease. Clin Neurol Neurosurg 2021; 208:106878. [PMID: 34418700 DOI: 10.1016/j.clineuro.2021.106878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether spiral analysis can monitor the effects of deep brain stimulation (DBS) in Parkinson disease (PD) and provide a window on clinical features that change post-operatively. Clinical evaluation after DBS is subjective and insensitive to small changes. Spiral analysis is a computerized test that quantifies kinematic, dynamic, and spatial aspects of spiral drawing. Validated computational indices are generated and correlate with a range of clinically relevant motor findings. These include measures of overall clinical severity (Severity), bradykinesia and rigidity (Smoothness), amount of tremor (Tremor), irregularity of drawing movements (Variability), and micrographia (Tightness). METHODS We retrospectively evaluated the effect of subthalamic nucleus (STN) (n = 66) and ventral intermediate thalamus (Vim) (n = 10) DBS on spiral drawing in PD subjects using spiral analysis. Subjects freely drew ten spirals on plain paper with an inking pen on a graphics tablet. Five spiral indices (Severity, Smoothness, Tremor, Variability, Tightness) were calculated and compared pre- and post-operatively using Wilcoxon-rank sum tests, adjusting for multiple comparisons. RESULTS Severity improved after STN and Vim DBS (p < 0.005). Smoothness (p < 0.01) and Tremor (p < 0.02) both improved after STN and Vim DBS. Variability improved only with Vim DBS. Neither STN nor Vim DBS significantly changed Tightness. CONCLUSIONS All major spiral indices, except Tightness, improved after DBS. This suggests spiral analysis monitors DBS effects in PD and provides an objective window on relevant clinical features that change post-operatively. It may thus have utilization in clinical trials or investigations into the neural pathways altered by DBS. The lack of change in Tightness supports the notion that DBS does not improve micrographia.
Collapse
Affiliation(s)
- Sara Radmard
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| | | | - Blair Ford
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nora Vanegas-Arroyave
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Guy M McKhann
- Department of Neurological Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Sameer A Sheth
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Linda Winfield
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Marta San Luciano
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | | | - Seth L Pullman
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
117
|
Lo Buono V, Lucà Trombetta M, Palmeri R, Bonanno L, Cartella E, Di Lorenzo G, Bramanti P, Marino S, Corallo F. Subthalamic nucleus deep brain stimulation and impulsivity in Parkinson's disease: a descriptive review. Acta Neurol Belg 2021; 121:837-847. [PMID: 33961279 PMCID: PMC8349322 DOI: 10.1007/s13760-021-01684-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/15/2021] [Indexed: 11/26/2022]
Abstract
Standard treatment of Parkinson’s disease involves the dopaminergic medications. Deep brain stimulation of the subthalamic nucleus (STN-DBS) is an important neurosurgical intervention often used as alternative treatment to drug therapy; however, it can be associated with increase of impulsive behaviors. This descriptive review focused on studies investigating the correlation between Deep brain stimulation of the subthalamic nucleus and impulsivity in Parkinson’s disease patients, arguing, the action’s mechanism and the specific role of the subthalamic nucleus. We searched on PubMed and Web of Science databases and screening references of included studies and review articles for additional citations. From initial 106 studies, only 15 met the search criteria. Parkinson’s Disease patients with and without Deep Brain Stimulation were compared with healthy controls, through 16 different tasks that assessed some aspects of impulsivity. Both Deep brain stimulation of the subthalamic nucleus and medication were associated with impulsive behavior and influenced decision-making processes. Moreover, findings demonstrated that: Impulse Control Disorders (ICDs) occurred soon after surgery, while, in pharmacological treatment, they appeared mainly after the initiation of treatment or the increase in dosage, especially with dopamine agonists. The subthalamic nucleus plays a part in the fronto-striato-thalamic-cortical loops mediating motor, cognitive, and emotional functions: this could explain the role of the Deep Brain Stimulation in behavior modulation in Parkinson’s Disease patients. Indeed, increase impulsivity has been reported also after deep brain stimulation of the subthalamic nucleus independently by dopaminergic medication status.
Collapse
Affiliation(s)
| | | | | | - Lilla Bonanno
- IRCCS Centro Neurolesi Bonino Pulejo, Messina, Italy
| | | | | | | | - Silvia Marino
- IRCCS Centro Neurolesi Bonino Pulejo, Messina, Italy
| | | |
Collapse
|
118
|
Chou KL, Charles D. Subthalamic Nucleus Deep Brain Stimulation: Uncomplicating Motor Complications for the Long Haul. Neurology 2021; 97:107-108. [PMID: 34078714 DOI: 10.1212/wnl.0000000000012245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 11/15/2022] Open
Affiliation(s)
- Kelvin L Chou
- From the Departments of Neurology and Neurosurgery (K.L.C.), University of Michigan, Ann Arbor; and Department of Neurology (D.C.), Vanderbilt University, Nashville, TN.
| | - David Charles
- From the Departments of Neurology and Neurosurgery (K.L.C.), University of Michigan, Ann Arbor; and Department of Neurology (D.C.), Vanderbilt University, Nashville, TN
| |
Collapse
|
119
|
Bove F, Mulas D, Cavallieri F, Castrioto A, Chabardès S, Meoni S, Schmitt E, Bichon A, Di Stasio E, Kistner A, Pélissier P, Chevrier E, Seigneuret E, Krack P, Fraix V, Moro E. Long-term Outcomes (15 Years) After Subthalamic Nucleus Deep Brain Stimulation in Patients With Parkinson Disease. Neurology 2021; 97:e254-e262. [PMID: 34078713 DOI: 10.1212/wnl.0000000000012246] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 04/13/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate the effects of deep brain stimulation of the subthalamic nucleus (STN-DBS) on motor complications in patients with Parkinson disease (PD) beyond 15 years after surgery. METHODS Data on motor complications, quality of life (QoL), activities of daily living, Unified Parkinson's Disease Rating Scale motor scores, dopaminergic treatment, stimulation measures, and side effects of STN-DBS were retrospectively retrieved and compared before surgery, at 1 year, and beyond 15 years after bilateral STN-DBS. RESULTS Fifty-one patients with 17.06 ± 2.18 years STN-DBS follow-up were recruited. Compared to baseline, the time spent with dyskinesia and the time spent in the "off" state were reduced by 75% (p < 0.001) and by 58.7% (p < 0.001), respectively. Moreover, dopaminergic drugs were reduced by 50.6% (p < 0.001). Parkinson's Disease Quality of Life Questionnaire total score and the emotional function and social function domains improved 13.8% (p = 0.005), 13.6% (p = 0.01), and 29.9% (p < 0.001), respectively. Few and mostly manageable device-related adverse events were observed during the follow-up. CONCLUSIONS STN-DBS is effective beyond 15 years from the intervention, notably with significant improvement in motor complications and stable reduction of dopaminergic drugs. Furthermore, despite the natural continuous progression of PD with worsening of levodopa-resistant motor and nonmotor symptoms over the years, patients undergoing STN-DBS could maintain an improvement in QoL. CLASSIFICATION OF EVIDENCE This study provides Class IV evidence that, for patients with PD, STN-DBS remains effective at treating motor complications 15 years after surgery.
Collapse
Affiliation(s)
- Francesco Bove
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Delia Mulas
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Francesco Cavallieri
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Anna Castrioto
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Stephan Chabardès
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Sara Meoni
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Emmanuelle Schmitt
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Amélie Bichon
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Enrico Di Stasio
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Andrea Kistner
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Pierre Pélissier
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Eric Chevrier
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Eric Seigneuret
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Paul Krack
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Valerie Fraix
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland
| | - Elena Moro
- From the Movement Disorders Unit, Division of Neurology (F.B., D.M., F.C., A.C., S.M., E.S., A.B., A.K., P.P., E.C., V.F., E.M.), CHU Grenoble Alpes, Grenoble, France; Neurology Unit (F.B.) and Chemistry, Biochemistry and Clinical Molecular Biology (E.D.S.), Fondazione Policlinico Universitario A. Gemelli IRCCS; Department of Neurosciences (F.B.) and Institute of Biochemistry and Clinical Biochemistry (E.D.S.), Università Cattolica del Sacro Cuore, Rome; Institute of Neurology (D.M.), Mater Olbia Hospital, Olbia; Neurology Unit, Neuromotor and Rehabilitation Department (F.C.), Azienda USL-IRCCS di Reggio Emilia; Clinical and Experimental Medicine PhD Program (F.C.), University of Modena and Reggio Emilia, Modena, Italy; Grenoble Institute of Neurosciences (A.C., S.C., S.M., E.S., A.B., A.K., P.P., E.C., E.S., V.F., E.M.), University Grenoble Alpes, Inserm, U1216, Grenoble; Division of Neurosurgery (S.C., E.S.), Centre Hospitalier Universitaire (CHU), Grenoble Alpes University, France; Department of Health Sciences (S.M.), University of Milan, Italy; and Department of Neurology (P.K.), Bern University Hospital, Switzerland.
| |
Collapse
|
120
|
Klink PC, Aubry JF, Ferrera VP, Fox AS, Froudist-Walsh S, Jarraya B, Konofagou EE, Krauzlis RJ, Messinger A, Mitchell AS, Ortiz-Rios M, Oya H, Roberts AC, Roe AW, Rushworth MFS, Sallet J, Schmid MC, Schroeder CE, Tasserie J, Tsao DY, Uhrig L, Vanduffel W, Wilke M, Kagan I, Petkov CI. Combining brain perturbation and neuroimaging in non-human primates. Neuroimage 2021; 235:118017. [PMID: 33794355 PMCID: PMC11178240 DOI: 10.1016/j.neuroimage.2021.118017] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Brain perturbation studies allow detailed causal inferences of behavioral and neural processes. Because the combination of brain perturbation methods and neural measurement techniques is inherently challenging, research in humans has predominantly focused on non-invasive, indirect brain perturbations, or neurological lesion studies. Non-human primates have been indispensable as a neurobiological system that is highly similar to humans while simultaneously being more experimentally tractable, allowing visualization of the functional and structural impact of systematic brain perturbation. This review considers the state of the art in non-human primate brain perturbation with a focus on approaches that can be combined with neuroimaging. We consider both non-reversible (lesions) and reversible or temporary perturbations such as electrical, pharmacological, optical, optogenetic, chemogenetic, pathway-selective, and ultrasound based interference methods. Method-specific considerations from the research and development community are offered to facilitate research in this field and support further innovations. We conclude by identifying novel avenues for further research and innovation and by highlighting the clinical translational potential of the methods.
Collapse
Affiliation(s)
- P Christiaan Klink
- Department of Vision & Cognition, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| | - Jean-François Aubry
- Physics for Medicine Paris, Inserm U1273, CNRS UMR 8063, ESPCI Paris, PSL University, Paris, France
| | - Vincent P Ferrera
- Department of Neuroscience & Department of Psychiatry, Columbia University Medical Center, New York, NY, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Andrew S Fox
- Department of Psychology & California National Primate Research Center, University of California, Davis, CA, USA
| | | | - Béchir Jarraya
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France; Foch Hospital, UVSQ, Suresnes, France
| | - Elisa E Konofagou
- Ultrasound and Elasticity Imaging Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Richard J Krauzlis
- Laboratory of Sensorimotor Research, National Eye Institute, Bethesda, MD, USA
| | - Adam Messinger
- Laboratory of Brain and Cognition, National Institute of Mental Health, Bethesda, MD, USA
| | - Anna S Mitchell
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom
| | - Michael Ortiz-Rios
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - Hiroyuki Oya
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Department of Neurosurgery, University of Iowa, Iowa city, IA, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, United Kingdom
| | - Anna Wang Roe
- Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou 310029, China
| | | | - Jérôme Sallet
- Department of Experimental Psychology, Oxford University, Oxford, United Kingdom; Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Michael Christoph Schmid
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom; Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, CH-1700 Fribourg, Switzerland
| | - Charles E Schroeder
- Nathan Kline Institute, Orangeburg, NY, USA; Columbia University, New York, NY, USA
| | - Jordy Tasserie
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Doris Y Tsao
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience; Howard Hughes Medical Institute; Computation and Neural Systems, Caltech, Pasadena, CA, USA
| | - Lynn Uhrig
- NeuroSpin, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de la Santé et de la Recherche Médicale (INSERM), Cognitive Neuroimaging Unit, Université Paris-Saclay, France
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, Neurosciences Department, KU Leuven Medical School, Leuven, Belgium; Leuven Brain Institute, KU Leuven, Leuven Belgium; Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital, Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| | - Melanie Wilke
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; Department of Cognitive Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Igor Kagan
- German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany.
| | - Christopher I Petkov
- Newcastle University Medical School, Newcastle upon Tyne NE1 7RU, United Kingdom.
| |
Collapse
|
121
|
Mitchell KT, Younce JR, Norris SA, Tabbal SD, Dowling JL, Rich KM, Perlmutter JS, Ushe M. Bilateral Subthalamic Nucleus Deep Brain Stimulation in Elderly Patients With Parkinson Disease: A Case-Control Study. Oper Neurosurg (Hagerstown) 2021; 19:234-240. [PMID: 32259239 DOI: 10.1093/ons/opaa049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/12/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Subthalamic nucleus deep brain stimulation (STN DBS) is an effective adjunctive therapy for Parkinson disease. Studies have shown improvement of motor function but often exclude patients older than 75 yr. OBJECTIVE To determine the safety and effectiveness of STN DBS in patients 75 yr and older. METHODS A total of 104 patients (52 patients >75 yr old, 52 patients <75 yr old) with STN DBS were paired and retrospectively analyzed. The primary outcome was change in Unified Parkinson Disease Rating Scale (UPDRS) subscale III at 1 yr postoperatively, OFF medication. Secondary outcomes were changes in UPDRS I, II, and IV subscales and levodopa equivalents. Complications and all-cause mortality were assessed at 30 d and 1 yr. RESULTS Both cohorts had significant improvements in UPDRS III at 6 mo and 1 yr with no difference between cohorts. Change in UPDRS III was noninferior to the younger cohort. The cohorts had similar worsening in UPDRS I at 1 yr, no change in UPDRS II, similar improvement in UPDRS IV, and similar levodopa equivalent reduction. There were similar numbers of postoperative intracerebral hemorrhages (2/52 in each cohort, more severe in the older cohort) and surgical complications (4/52 in each cohort), and mortality in the older cohort was similar to an additional matched cohort not receiving DBS. CONCLUSION STN DBS provides substantial motor benefit and reduction in levodopa equivalents with a low rate of complications in older patients, which is also noninferior to the benefit in younger patients. STN DBS remains an effective therapy for those over 75 yr.
Collapse
Affiliation(s)
- Kyle T Mitchell
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - John R Younce
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Scott A Norris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Samer D Tabbal
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Department of Neurology, American University of Beirut, Beirut, Lebanon
| | - Joshua L Dowling
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Keith M Rich
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Joel S Perlmutter
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri.,Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri.,Program in Physical Therapy, Washington University School of Medicine, St. Louis, Missouri.,Program in Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri
| | - Mwiza Ushe
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
122
|
Wenzel GR, Roediger J, Brücke C, Marcelino ALDA, Gülke E, Pötter-Nerger M, Scholtes H, Wynants K, Juárez Paz LM, Kühn AA. CLOVER-DBS: Algorithm-Guided Deep Brain Stimulation-Programming Based on External Sensor Feedback Evaluated in a Prospective, Randomized, Crossover, Double-Blind, Two-Center Study. JOURNAL OF PARKINSONS DISEASE 2021; 11:1887-1899. [PMID: 34151855 DOI: 10.3233/jpd-202480] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent technological advances in deep brain stimulation (DBS) (e.g., directional leads, multiple independent current sources) lead to increasing DBS-optimization burden. Techniques to streamline and facilitate programming could leverage these innovations. OBJECTIVE We evaluated clinical effectiveness of algorithm-guided DBS-programming based on wearable-sensor-feedback compared to standard-of-care DBS-settings in a prospective, randomized, crossover, double-blind study in two German DBS centers. METHODS For 23 Parkinson's disease patients with clinically effective DBS, new algorithm-guided DBS-settings were determined and compared to previously established standard-of-care DBS-settings using UPDRS-III and motion-sensor-assessment. Clinical and imaging data with lead-localizations were analyzed to evaluate characteristics of algorithm-derived programming compared to standard-of-care. Six different versions of the algorithm were evaluated during the study and 10 subjects programmed with uniform algorithm-version were analyzed as a subgroup. RESULTS Algorithm-guided and standard-of-care DBS-settings effectively reduced motor symptoms compared to off-stimulation-state. UPDRS-III scores were reduced significantly more with standard-of-care settings as compared to algorithm-guided programming with heterogenous algorithm versions in the entire cohort. A subgroup with the latest algorithm version showed no significant differences in UPDRS-III achieved by the two programming-methods. Comparing active contacts in standard-of-care and algorithm-guided DBS-settings, contacts in the latter had larger location variability and were farther away from a literature-based optimal stimulation target. CONCLUSION Algorithm-guided programming may be a reasonable approach to replace monopolar review, enable less trained health-professionals to achieve satisfactory DBS-programming results, or potentially reduce time needed for programming. Larger studies and further improvements of algorithm-guided programming are needed to confirm these results.
Collapse
Affiliation(s)
- Gregor R Wenzel
- Department of Neurology, Movement disorders & Neuromodulation section, Charité -University Medicine Berlin, Germany
| | - Jan Roediger
- Department of Neurology, Movement disorders & Neuromodulation section, Charité -University Medicine Berlin, Germany.,Einstein Center for Neurosciences Berlin, Charité -University Medicine Berlin, Germany
| | - Christof Brücke
- Department of Neurology, Movement disorders & Neuromodulation section, Charité -University Medicine Berlin, Germany
| | - Ana Luísa de A Marcelino
- Department of Neurology, Movement disorders & Neuromodulation section, Charité -University Medicine Berlin, Germany
| | - Eileen Gülke
- Department of Neurology, Universitätsklinikum Hamburg-Eppendorf, Germany
| | | | | | | | | | - Andrea A Kühn
- Department of Neurology, Movement disorders & Neuromodulation section, Charité -University Medicine Berlin, Germany
| |
Collapse
|
123
|
Zeugin D, Ionta S. Anatomo-Functional Origins of the Cortical Silent Period: Spotlight on the Basal Ganglia. Brain Sci 2021; 11:705. [PMID: 34071742 PMCID: PMC8227635 DOI: 10.3390/brainsci11060705] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
The so-called cortical silent period (CSP) refers to the temporary interruption of electromyographic signal from a muscle following a motor-evoked potential (MEP) triggered by transcranial magnetic stimulation (TMS) over the primary motor cortex (M1). The neurophysiological origins of the CSP are debated. Previous evidence suggests that both spinal and cortical mechanisms may account for the duration of the CSP. However, contextual factors such as cortical fatigue, experimental procedures, attentional load, as well as neuropathology can also influence the CSP duration. The present paper summarizes the most relevant evidence on the mechanisms underlying the duration of the CSP, with a particular focus on the central role of the basal ganglia in the "direct" (excitatory), "indirect" (inhibitory), and "hyperdirect" cortico-subcortical pathways to manage cortical motor inhibition. We propose new methods of interpretation of the CSP related, at least partially, to the inhibitory hyperdirect and indirect pathways in the basal ganglia. This view may help to explain the respective shortening and lengthening of the CSP in various neurological disorders. Shedding light on the complexity of the CSP's origins, the present review aims at constituting a reference for future work in fundamental research, technological development, and clinical settings.
Collapse
Affiliation(s)
| | - Silvio Ionta
- Sensory-Motor Laboratory (SeMoLa), Jules-Gonin Eye Hospital/Fondation Asile des Aveugles, Department of Ophthalmology, University of Lausanne, 1002 Lausanne, Switzerland
| |
Collapse
|
124
|
Schnitzler A, Mir P, Brodsky MA, Verhagen L, Groppa S, Alvarez R, Evans A, Blazquez M, Nagel S, Pilitsis JG, Pötter-Nerger M, Tse W, Almeida L, Tomycz N, Jimenez-Shahed J, Libionka W, Carrillo F, Hartmann CJ, Groiss SJ, Glaser M, Defresne F, Karst E, Cheeran B, Vesper J. Directional Deep Brain Stimulation for Parkinson's Disease: Results of an International Crossover Study With Randomized, Double-Blind Primary Endpoint. Neuromodulation 2021; 25:817-828. [PMID: 34047410 DOI: 10.1111/ner.13407] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Published reports on directional deep brain stimulation (DBS) have been limited to small, single-center investigations. Therapeutic window (TW) is used to describe the range of stimulation amplitudes achieving symptom relief without side effects. This crossover study performed a randomized double-blind assessment of TW for directional and omnidirectional DBS in a large cohort of patients implanted with a DBS system in the subthalamic nucleus for Parkinson's disease. MATERIALS AND METHODS Participants received omnidirectional stimulation for the first three months after initial study programming, followed by directional DBS for the following three months. The primary endpoint was a double-blind, randomized evaluation of TW for directional vs. omnidirectional stimulation at three months after initial study programming. Additional data recorded at three- and six-month follow-ups included stimulation preference, therapeutic current strength, Unified Parkinson's Disease Rating Scale (UPDRS) part III motor score, and quality of life. RESULTS The study enrolled 234 subjects (62 ± 8 years, 33% female). TW was wider using directional stimulation in 183 of 202 subjects (90.6%). The mean increase in TW with directional stimulation was 41% (2.98 ± 1.38 mA, compared to 2.11 ± 1.33 mA for omnidirectional). UPDRS part III motor score on medication improved 42.4% at three months (after three months of omnidirectional stimulation) and 43.3% at six months (after three months of directional stimulation) with stimulation on, compared to stimulation off. After six months, 52.8% of subjects blinded to stimulation type (102/193) preferred the period with directional stimulation, and 25.9% (50/193) preferred the omnidirectional period. The directional period was preferred by 58.5% of clinicians (113/193) vs. 21.2% (41/193) who preferred the omnidirectional period. CONCLUSION Directional stimulation yielded a wider TW compared to omnidirectional stimulation and was preferred by blinded subjects and clinicians.
Collapse
Affiliation(s)
- Alfons Schnitzler
- Department of Neurology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Pablo Mir
- Clinical Neurology and Neurophysiology Department, Movement Disorders Unit, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital, CSIC/University of Seville, Seville, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Seville, Spain
| | - Matthew A Brodsky
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Leonard Verhagen
- Department of Neurological Sciences, Rush University, Chicago, IL, USA
| | - Sergiu Groppa
- Johannes Gutenberg University of Mainz, Clinic of Neurology, Mainz, Germany
| | - Ramiro Alvarez
- Department of Neurology, Hospital Trias i Pujol, Badalona, Spain
| | - Andrew Evans
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Australia
| | - Marta Blazquez
- Department of Neurology, Hospital Universitario Central de Asturias, Spain
| | - Sean Nagel
- Department of Neurosurgery, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Julie G Pilitsis
- Department of Neurosurgery, Albany Medical Center, New York, NY, USA
| | | | - Winona Tse
- Department of Neurology, Mount Sinai Hospital, New York, NY, USA
| | - Leonardo Almeida
- Department of Neurology, Shands at University of Florida, Gainesville, FL, USA
| | - Nestor Tomycz
- Department of Neurosurgery, Allegheny General Hospital, Pittsburgh, PA, USA
| | | | - Witold Libionka
- Department of Neurology, Copernicus Hospital, Gdansk, Poland
| | - Fatima Carrillo
- Clinical Neurology and Neurophysiology Department, Movement Disorders Unit, Institute of Biomedicine of Seville, Virgen del Rocío University Hospital, CSIC/University of Seville, Seville, Spain
| | - Christian J Hartmann
- Department of Neurology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Stefan Jun Groiss
- Department of Neurology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Martin Glaser
- Department of Neurosurgery, Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | - Edward Karst
- Abbott, Medical and Clinical Affairs, Plano, TX, USA
| | | | - Jan Vesper
- Department of Neurosurgery, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | | |
Collapse
|
125
|
Milosevic L, Kalia SK, Hodaie M, Lozano AM, Popovic MR, Hutchison WD, Lankarany M. A theoretical framework for the site-specific and frequency-dependent neuronal effects of deep brain stimulation. Brain Stimul 2021; 14:807-821. [PMID: 33991712 DOI: 10.1016/j.brs.2021.04.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/01/2021] [Accepted: 04/27/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Deep brain stimulation is an established therapy for several neurological disorders; however, its effects on neuronal activity vary across brain regions and depend on stimulation settings. Understanding these variable responses can aid in the development of physiologically-informed stimulation paradigms in existing or prospective indications. OBJECTIVE Provide experimental and computational insights into the brain-region-specific and frequency-dependent effects of extracellular stimulation on neuronal activity. METHODS In patients with movement disorders, single-neuron recordings were acquired from the subthalamic nucleus, substantia nigra pars reticulata, ventral intermediate nucleus, or reticular thalamus during microstimulation across various frequencies (1-100 Hz) to assess single-pulse and frequency-response functions. Moreover, a biophysically-realistic computational framework was developed which generated postsynaptic responses under the assumption that electrical stimuli simultaneously activated all convergent presynaptic inputs to stimulation target neurons. The framework took into consideration the relative distributions of excitatory/inhibitory afferent inputs to model site-specific responses, which were in turn embedded within a model of short-term synaptic plasticity to account for stimulation frequency-dependence. RESULTS We demonstrated microstimulation-evoked excitatory neuronal responses in thalamic structures (which have predominantly excitatory inputs) and inhibitory responses in basal ganglia structures (predominantly inhibitory inputs); however, higher stimulation frequencies led to a loss of site-specificity and convergence towards neuronal suppression. The model confirmed that site-specific responses could be simulated by accounting for local neuroanatomical/microcircuit properties, while suppression of neuronal activity during high-frequency stimulation was mediated by short-term synaptic depression. CONCLUSIONS Brain-region-specific and frequency-dependant neuronal responses could be simulated by considering neuroanatomical (local microcircuitry) and neurophysiological (short-term plasticity) properties.
Collapse
Affiliation(s)
- Luka Milosevic
- Krembil Brain Institute, University Health Network, Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada; CRANIA, University Health Network and University of Toronto, Toronto, Canada.
| | - Suneil K Kalia
- Krembil Brain Institute, University Health Network, Toronto, Canada; KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada; CRANIA, University Health Network and University of Toronto, Toronto, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada; Department of Surgery, University of Toronto, Toronto, Canada
| | - Mojgan Hodaie
- Krembil Brain Institute, University Health Network, Toronto, Canada; CRANIA, University Health Network and University of Toronto, Toronto, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada; Department of Surgery, University of Toronto, Toronto, Canada
| | - Andres M Lozano
- Krembil Brain Institute, University Health Network, Toronto, Canada; CRANIA, University Health Network and University of Toronto, Toronto, Canada; Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Canada; Department of Surgery, University of Toronto, Toronto, Canada
| | - Milos R Popovic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada; CRANIA, University Health Network and University of Toronto, Toronto, Canada
| | - William D Hutchison
- CRANIA, University Health Network and University of Toronto, Toronto, Canada; Department of Surgery, University of Toronto, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Milad Lankarany
- Krembil Brain Institute, University Health Network, Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; KITE, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
| |
Collapse
|
126
|
Liu C, Lee CY, Asher G, Cao L, Terakoshi Y, Cao P, Kobayakawa R, Kobayakawa K, Sakurai K, Liu Q. Posterior subthalamic nucleus (PSTh) mediates innate fear-associated hypothermia in mice. Nat Commun 2021; 12:2648. [PMID: 33976193 PMCID: PMC8113537 DOI: 10.1038/s41467-021-22914-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 03/17/2021] [Indexed: 12/16/2022] Open
Abstract
The neural mechanisms of fear-associated thermoregulation remain unclear. Innate fear odor 2-methyl-2-thiazoline (2MT) elicits rapid hypothermia and elevated tail temperature, indicative of vasodilation-induced heat dissipation, in wild-type mice, but not in mice lacking Trpa1-the chemosensor for 2MT. Here we report that Trpa1-/- mice show diminished 2MT-evoked c-fos expression in the posterior subthalamic nucleus (PSTh), external lateral parabrachial subnucleus (PBel) and nucleus of the solitary tract (NTS). Whereas tetanus toxin light chain-mediated inactivation of NTS-projecting PSTh neurons suppress, optogenetic activation of direct PSTh-rostral NTS pathway induces hypothermia and tail vasodilation. Furthermore, selective opto-stimulation of 2MT-activated, PSTh-projecting PBel neurons by capturing activated neuronal ensembles (CANE) causes hypothermia. Conversely, chemogenetic suppression of vGlut2+ neurons in PBel or PSTh, or PSTh-projecting PBel neurons attenuates 2MT-evoked hypothermia and tail vasodilation. These studies identify PSTh as a major thermoregulatory hub that connects PBel to NTS to mediate 2MT-evoked innate fear-associated hypothermia and tail vasodilation.
Collapse
Affiliation(s)
- Can Liu
- Peking University-Tsinghua University-NIBS Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, China
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Chia-Ying Lee
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Greg Asher
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Liqin Cao
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuka Terakoshi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Peng Cao
- National Institute of Biological Sciences (NIBS), Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China
| | - Reiko Kobayakawa
- Department of Functional Neuroscience, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Ko Kobayakawa
- Department of Functional Neuroscience, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Katsuyasu Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Qinghua Liu
- National Institute of Biological Sciences (NIBS), Beijing, China.
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Tsinghua University, Beijing, China.
| |
Collapse
|
127
|
Martínez-Fernández R, Matarazzo M, Máñez-Miró JU, Obeso JA. The Role of Focused Ultrasound in the Management of Movement Disorders: Insights after 5 Years of Experience. Mov Disord Clin Pract 2021; 8:681-687. [PMID: 34307739 DOI: 10.1002/mdc3.13223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/07/2021] [Indexed: 11/07/2022] Open
Affiliation(s)
- Raúl Martínez-Fernández
- HM CINAC (Centro Integral de Neurociencias Abarca Campal) Hospital Universitario HM Puerta del Sur, HM Hospitales Madrid Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases Carlos III Institute Madrid Spain
| | - Michele Matarazzo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal) Hospital Universitario HM Puerta del Sur, HM Hospitales Madrid Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases Carlos III Institute Madrid Spain
| | - Jorge U Máñez-Miró
- HM CINAC (Centro Integral de Neurociencias Abarca Campal) Hospital Universitario HM Puerta del Sur, HM Hospitales Madrid Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases Carlos III Institute Madrid Spain
| | - Jose A Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal) Hospital Universitario HM Puerta del Sur, HM Hospitales Madrid Spain.,Network Center for Biomedical Research on Neurodegenerative Diseases Carlos III Institute Madrid Spain
| |
Collapse
|
128
|
Liu J, Ding H, Xu K, Liu R, Wang D, Ouyang J, Liu Z, Miao Z. Pallidal versus subthalamic deep-brain stimulation for meige syndrome: a retrospective study. Sci Rep 2021; 11:8742. [PMID: 33888857 PMCID: PMC8062505 DOI: 10.1038/s41598-021-88384-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/12/2021] [Indexed: 11/09/2022] Open
Abstract
Deep-brain stimulation (DBS) is an effective treatment for patients with Meige syndrome. The globus pallidus interna (GPi) and the subthalamic nucleus (STN) are accepted targets for this treatment. We compared 12-month outcomes for patients who had undergone bilateral stimulation of the GPi or STN. Forty-two Asian patients with primary Meige syndrome who underwent GPi or STN neurostimulation were recruited between September 2017 and September 2019 at the Department of Neurosurgery, Peking University People's Hospital. The primary outcome was the change in motor function, including the Burke-Fahn-Marsden Dystonia Rating Scale movement (BFMDRS-M) and disability subscale (BFMDRS-D) at 3 days before DBS (baseline) surgery and 1, 3, 6, and 12 months after surgery. Secondary outcomes included health-related quality of life, sleep quality status, depression severity, and anxiety severity at 3 days before and 12 months after DBS surgery. Adverse events during the 12 months were also recorded. Changes in BFMDRS-M and BFMDRS-D scores at 1, 3, 6, and 12 months with DBS and without medication did not significantly differ based on the stimulation target. There were also no significant differences in the changes in health-related quality of life (36-Item Short-Form General Health Survey) and sleep quality status (Pittsburgh Sleep Quality Index) at 12 months. However, there were larger improvements in the STN than the GPi group in mean score changes on the 17-item Hamilton depression rating scale (- 3.38 vs. - 0.33 points; P = 0.014) and 14-item Hamilton anxiety rating scale (- 3.43 vs. - 0.19 points; P < 0.001). There were no significant between-group differences in the frequency or type of serious adverse events. Patients with Meige syndrome had similar improvements in motor function, quality of life and sleep after either pallidal or subthalamic stimulation. Depression and anxiety factors may reasonably be included during the selection of DBS targets for Meige syndrome.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Hu Ding
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Ke Xu
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Ruen Liu
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China.
| | - Dongliang Wang
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Jia Ouyang
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Zhi Liu
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| | - Zeyu Miao
- Department of Neurosurgery, Peking University People's Hospital, 11th Xizhimen South St., Beijing, 100044, China
| |
Collapse
|
129
|
Costanza A, Radomska M, Bondolfi G, Zenga F, Amerio A, Aguglia A, Serafini G, Amore M, Berardelli I, Pompili M, Nguyen KD. Suicidality Associated With Deep Brain Stimulation in Extrapyramidal Diseases: A Critical Review and Hypotheses on Neuroanatomical and Neuroimmune Mechanisms. Front Integr Neurosci 2021; 15:632249. [PMID: 33897384 PMCID: PMC8060445 DOI: 10.3389/fnint.2021.632249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Deep brain stimulation (DBS) is a very well-established and effective treatment for patients with extrapyramidal diseases. Despite its generally favorable clinical efficacy, some undesirable outcomes associated with DBS have been reported. Among such complications are incidences of suicidal ideation (SI) and behavior (SB) in patients undergoing this neurosurgical procedure. However, causal associations between DBS and increased suicide risk are not demonstrated and they constitute a debated issue. In light of these observations, the main objective of this work is to provide a comprehensive and unbiased overview of the literature on suicide risk in patients who received subthalamic nucleus (STN) and internal part of globus pallidum (GPi) DBS treatment. Additionally, putative mechanisms that might be involved in the development of SI and SB in these patients as well as caveats associated with these hypotheses are introduced. Finally, we briefly propose some clinical implications, including therapeutic strategies addressing these potential disease mechanisms. While a mechanistic connection between DBS and suicidality remains a controversial topic that requires further investigation, it is of critical importance to consider suicide risk as an integral component of candidate selection and post-operative care in DBS.
Collapse
Affiliation(s)
- Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, ASO Santi Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| | - Michalina Radomska
- Faculty of Psychology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Guido Bondolfi
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, Service of Liaison Psychiatry and Crisis Intervention (SPLIC), Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Francesco Zenga
- Department of Neurosurgery, University and City of Health and Science Hospital, Turin, Italy
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Mood Disorders Program, Tufts Medical Center, Boston, MA, United States
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Isabella Berardelli
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Khoa D Nguyen
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, United States.,Tranquis Therapeutics, Palo Alto, CA, United States.,Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
130
|
Park E, Lyon JG, Alvarado-Velez M, Betancur MI, Mokarram N, Shin JH, Bellamkonda RV. Enriching neural stem cell and anti-inflammatory glial phenotypes with electrical stimulation after traumatic brain injury in male rats. J Neurosci Res 2021; 99:1864-1884. [PMID: 33772860 PMCID: PMC8360147 DOI: 10.1002/jnr.24834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) by an external physical impact results in compromised brain function via undesired neuronal death. Following the injury, resident and peripheral immune cells, astrocytes, and neural stem cells (NSCs) cooperatively contribute to the recovery of the neuronal function after TBI. However, excessive pro‐inflammatory responses of immune cells, and the disappearance of endogenous NSCs at the injury site during the acute phase of TBI, can exacerbate TBI progression leading to incomplete healing. Therefore, positive outcomes may depend on early interventions to control the injury‐associated cellular milieu in the early phase of injury. Here, we explore electrical stimulation (ES) of the injury site in a rodent model (male Sprague–Dawley rats) to investigate its overall effect on the constituent brain cell phenotype and composition during the acute phase of TBI. Our data showed that a brief ES for 1 hr on day 2 of TBI promoted anti‐inflammatory phenotypes of microglia as assessed by CD206 expression and increased the population of NSCs and Nestin+ astrocytes at 7 days post‐TBI. Also, ES effectively increased the number of viable neurons when compared to the unstimulated control group. Given the salience of microglia and neural stem cells for healing after TBI, our results strongly support the potential benefit of the therapeutic use of ES during the acute phase of TBI to regulate neuroinflammation and to enhance neuroregeneration.
Collapse
Affiliation(s)
- Eunyoung Park
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Melissa Alvarado-Velez
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha I Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Nassir Mokarram
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Ravi V Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
131
|
Zhang F, Wang F, Li W, Wang N, Han C, Fan S, Li P, Xu L, Zhang J, Meng F. Relationship between electrode position of deep brain stimulation and motor symptoms of Parkinson's disease. BMC Neurol 2021; 21:122. [PMID: 33731033 PMCID: PMC7972210 DOI: 10.1186/s12883-021-02148-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Background To investigate the relationship between the position of bilateral STN-DBS location of active contacts and the clinical efficacy of STN-DBS on motor symptoms in Parkinson’s disease (PD) patients. Methods Retrospectively analyze the clinical data of 57 patients with PD who underwent bilateral STN-DBS from March 2018 to December 2018. Unified Parkinson’s Disease Rating Scale-Part III (UPDRS-III) score, levodopa equivalent day dose (LEDD), Parkinson’s Disease Quality of Life Scale (PDQ-39) before operation and within 6 months after operation, determine the location of activated contacts and volume of tissue activated (VTA) in the Montreal Neurological Institute (MNI) space, and analyze their correlation with the improvement rate of motor symptoms (UPDRS-III score improvement rate). Results After 6 months of follow up, the UPDRS-III scores of 57 patients (Med-off) were improved by 55.4 ± 18.9% (P<0.001) compared with that before operation. The improvement rate of PDQ-39 scores [(47.4 ± 23.2)%, (P < 0.001)] and the reduction rate of LEDD [(40.1 ± 24.3)%, (P < 0.01)] at 6 months postoperation were positively correlated with the improvement rate of motor symptoms (Med-off)(PDQ-39:r = 0.461, P<0.001; LEDD: r = 0.354, P = 0.007), the improvement rate of UPDRS-III (Med-off) and the Z-axis coordinate of the active contact in the MNI space were positively correlated (left side: r = 0.349,P = 0.008;right side: r = 0.369,P = 0.005). In the MNI space, there was no correlation between the UPDRS-III scores improvement rate (Med-off) at 6 months after operation and bilateral VTA in the STN motor subregion, STN associative subregion and STN limbic subregion of the active electrode contacts of 57 patients (all P > 0.05). At 6 months after surgery, the difference between the Z-axis coordinate in the different improvement rate subgroups(<25, 25 to 50%, and>50%) in the MNI space was statistically significant (left side: P = 0.030; right side: P = 0.024). In the MNI space, there was no statistically significant difference between the groups in the VTA of the electrode active contacts (all P > 0.05). Conclusion STN-DBS can improve the motor symptoms of PD patients and improve the quality of life. The closer the stimulation is to the STN dorsolateral sensorimotor area, the higher the DBS is to improve the motor symptoms of PD patients.
Collapse
Affiliation(s)
- Feng Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.,Department of neurosurgery, the First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Feng Wang
- Departments of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, 310000, Hangzhou, China
| | - Weiguo Li
- Department of neurosurgery, QiLu Hospital of Shandong University, Jinan, 250012, China
| | - Ning Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Chunlei Han
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Shiying Fan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Peng Li
- Department of neurosurgery, the First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Lifeng Xu
- Department of neurosurgery, the First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China
| | - Jianguo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Fangang Meng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China. .,Beijing Key Laboratory of Neurostimulation, Beijing, 100070, China. .,Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
132
|
Kwan V, Shum D, Haffenden A, Yeates KO, Kwok A, Lau H, Poon WS, Chan D, Zhu XL, Chan D, Mok V, Chan A, Ma K, Yeung J, Lau C, Bezchlibnyk Y, Kiss Z, Tang V. A retrospective comparison of cognitive performance in individuals with advanced Parkinson's Disease in Hong Kong and Canada. APPLIED NEUROPSYCHOLOGY-ADULT 2021; 29:1562-1570. [PMID: 33721508 DOI: 10.1080/23279095.2021.1898396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A deeper understanding of the cross-cultural applicability of cognitive tests across countries and cultures is needed to better equip neuropsychologists for the assessment of patients from diverse backgrounds. Our study compared cognitive test scores in patients with advanced Parkinson's disease (PD) at the Prince of Wales Hospital (n = 63; Hong Kong) and the Foothills Medical Center (n = 20; Calgary, Canada). The groups did not differ in age or sex (p > .05), but Western patients had significantly more years of education (M = 14.2, SD = 2.7) than Asian patients (M = 10.33, SD = 4.4). Cognitive tests administered to both groups included: digit span, verbal fluency (animals), the Boston Naming Test, and verbal memory (California Verbal Learning Test or Chinese Auditory Verbal Learning Test). Testing was completed before and 12 months after deep brain stimulation surgery. Results showed cognitive performance was similar across time, but significant group differences were found on digit span forward (longer among patients from Hong Kong; F(1, 75) = 44.155, p < .001) and the Boston Naming Test (higher percent spontaneous correct among patients from Canada; F(1, 62) = 7.218, p = .009, η2 = 0.104), after controlling for age, sex, and years of education. In conclusion, our findings provide preliminary support for the similarity of Chinese versions of tests originally developed for Western populations. Also, we caution that some aspects of testing may be susceptible to cultural bias and therefore warrant attention in clinical practice and refinement in future test development for Asian patients.
Collapse
Affiliation(s)
| | - David Shum
- The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | | | | | - Alice Kwok
- Department of Clinical Psychology, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Herman Lau
- Chinese University Medical Centre, Ma Liu Shui, Hong Kong
| | - Wai Sang Poon
- Department of Surgery, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Danny Chan
- Department of Surgery, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - X L Zhu
- Department of Surgery, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - David Chan
- Department of Surgery, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Vincent Mok
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Anne Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Karen Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Jonas Yeung
- Department of Medicine, Alice Ho Miu Ling Nethersole Hospital, Tai Po, Hong Kong
| | - Claire Lau
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Sha Tin, Hong Kong
| | - Yarema Bezchlibnyk
- Department of Clinical Neurosciences, University of Calgary, Alberta, Canada
| | - Zelma Kiss
- Department of Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Venus Tang
- Department of Clinical Psychology, Prince of Wales Hospital, Sha Tin, Hong Kong
| |
Collapse
|
133
|
Brunet J, Price J, Wurz A, McDonough M, Nantel J. Boxing with Parkinson's Disease: findings from a qualitative study using self-determination theory. Disabil Rehabil 2021; 44:3880-3889. [PMID: 33625954 DOI: 10.1080/09638288.2021.1891465] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Adults with Parkinson's disease (PD) experience debilitating symptoms that may be mitigated by boxing. Yet, attrition from boxing programs is problematic. Understanding whether PD-specific boxing programs can enhance motivation to remain physically active is important. This study investigated adults' experiences within a PD-specific boxing program and explored their perspectives on how the program has influenced physical activity motivational processes using a self-determination theory (SDT) lens. Methods: Nine adults with PD who took part in the Boxing 4 Health PD-specific boxing program completed face-to-face, semi-structured interviews. Data were analyzed using a hybrid approach of inductive and deductive thematic analysis. Results: The analysis yielded five themes: (1) I made an informed decision to participate and it feels good, (2) Boxing helps me stay active and keep moving, (3) To keep me going, the boxing program needs to have variety and allow for input, (4) A program that can be adapted to me is important, and (5) The instructor and the group facilitate my continued participation in the program. Conclusion: Providing participants with adapted exercises, varied sessions, and opportunities to provide input in a group-based PD-specific boxing program may be conducive to enhancing motivation for physical activity. Further, SDT may be a useful theoretical framework for developing and evaluating PD-specific programs.IMPLICATIONS FOR REHABILITATIONAdults with Parkinson's disease (PD) have many reasons for not engaging in physical activity, including a lack of motivation.A group-based PD-specific boxing program may be conducive to increasing motivation for physical activity.Professionals should consider: (1) educating adults with PD on the benefits of boxing, (2) offering group-based PD-specific boxing programs that are adaptable, varied, and open to input, and (3) fostering social support and networking opportunities within such programs.
Collapse
Affiliation(s)
- Jennifer Brunet
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, Canada.,Cancer Therapeutic Program, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Canada.,Institut du savoir Montfort, Hôpital Montfort, Ottawa, Canada
| | - Jenson Price
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| | - Amanda Wurz
- Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | | | - Julie Nantel
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, Canada
| |
Collapse
|
134
|
Sadighi M, Şişman M, Açıkgöz BC, Eroğlu HH, Eyüboğlu BM. Low-frequency conductivity tensor imaging with a single current injection using DT-MREIT. Phys Med Biol 2021; 66:055011. [PMID: 33472190 DOI: 10.1088/1361-6560/abddcf] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Diffusion tensor-magnetic resonance electrical impedance tomography (DT-MREIT) is an imaging modality to obtain low-frequency anisotropic conductivity distribution employing diffusion tensor imaging and MREIT techniques. DT-MREIT is based on the linear relationship between the conductivity and water self-diffusion tensors in a porous medium, like the brain white matter. Several DT-MREIT studies in the literature provide cross-sectional anisotropic conductivity images of tissue phantoms, canine brain, and the human brain. In these studies, the conductivity tensor images are reconstructed using the diffusion tensor and current density data acquired by injecting two linearly independent current patterns. In this study, a novel reconstruction algorithm is devised for DT-MREIT to reconstruct the conductivity tensor images using a single current injection. Therefore, the clinical applicability of DT-MREIT can be improved by reducing the total acquisition time, the number of current injection cables, and contact electrodes to half by decreasing the number of current injection patterns to one. The proposed method is evaluated utilizing simulated measurements and physical experiments. The results obtained show the successful reconstruction of the anisotropic conductivity distribution using the proposed single current DT-MREIT.
Collapse
Affiliation(s)
- Mehdi Sadighi
- Department of Electrical and Electronics Engineering, Middle East Technical University, 06800 Ankara, Turkey
| | | | | | | | | |
Collapse
|
135
|
Apetz N, Paralikar K, Neumaier B, Drzezga A, Wiedermann D, Iyer R, Munns G, Scott E, Timmermann L, Endepols H. Towards chronic deep brain stimulation in freely moving hemiparkinsonian rats: Applicability and functionality of a fully implantable stimulation system. J Neural Eng 2021; 18. [PMID: 33607640 DOI: 10.1088/1741-2552/abe806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/19/2021] [Indexed: 12/23/2022]
Abstract
Objective This study aimed at investigating a novel fully implantable deep brain stimulation system and its ability to modulate brain metabolism and behavior through subthalamic nucleus stimulation in a hemiparkinsonian rat model. Approach Twelve male rats were unilaterally lesioned with 6-hydroxydopamine in the medial forebrain bundle and received a fully implantable deep brain stimulation system aiming at the ipsilesional subthalamic nucleus. Each rat underwent three cylinder tests to analyze front paw use: A PRE test before any surgical intervention, an OFF test after surgery but before stimulation onset and an ON test under deep brain stimulation. To visualize brain glucose metabolism in the awake animal, two [18F]FDG scans were conducted in the OFF and ON condition. At least four weeks after surgery, an [18F]FDOPA scan was used to check for dopaminergic integrity. Main results In general, STN DBS increased [18F]FDG uptake ipsilesionally and decreased it contralesionally. More specifically, bilateral orbitofrontal cortex, ipsilateral caudate putamen, sensorimotor cortex and nucleus accumbens showed significantly higher tracer uptake in ON compared to OFF condition. Contralateral cingulate and secondary motor cortex, caudate putamen, amygdala, hippocampus, retrosplenial granular cortex, superior colliculus, and parts of the cerebellum exhibited significantly higher [18F]FDG uptake in the OFF condition. On the behavioral level, stimulation was able improve use of the contralesional affected front paw suggesting an effective stimulation produced by the implanted system. Significance The fully implantable stimulation system developed by us and presented here offers the output of arbitrary user-defined waveforms, patterns and stimulation settings and allows tracer accumulation in freely moving animals. It is therefore a suitable device for implementing behavioral PET studies. It contributes immensely to the possibilities to characterize and unveil the effects and mechanisms of deep brain stimulation offering valuable clues for future improvements of this therapy.
Collapse
Affiliation(s)
- Nadine Apetz
- Institute of Radiochemistry and Experimental Molegular Imaging, University Hospital Cologne, Kerpener Str. 62, Koln, Nordrhein-Westfalen, 50937, GERMANY
| | - Kunal Paralikar
- Medtronic Inc, 7000 Central Avenue NE Friedley, Minneapolis, Minnesota, 55432-5604, UNITED STATES
| | - Bernd Neumaier
- Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, Julich, Nordrhein-Westfalen, 52428, GERMANY
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Kerpener Str. 62, Koln, Nordrhein-Westfalen, 50937, GERMANY
| | - Dirk Wiedermann
- Multimodal Imaging Group, Max Planck Institute for Metabolism Research, Gleueler Str. 50, Köln, 50931, GERMANY
| | - Rajesh Iyer
- Medtronic Inc, 7000 Central Avenue NE Fridley, Minneapolis, Minnesota, 55432-5604, UNITED STATES
| | - Gordon Munns
- Medtronic Inc, 7000 Central Avenue NE Friedley, Minneapolis, Minnesota, 55432-5604, UNITED STATES
| | - Erik Scott
- Medtronic Inc, 7000 Central Avenue NE Friedley, Minneapolis, Minnesota, 55432-5604, UNITED STATES
| | - Lars Timmermann
- Department of Neurology, University Hospital Marburg Center of Neurology, Baldingerstraße, Marburg, Hessen, 35039, GERMANY
| | - Heike Endepols
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital Cologne, Kerpener Str. 62, Koln, Nordrhein-Westfalen, 50937, GERMANY
| |
Collapse
|
136
|
Timing variability and midfrontal ~4 Hz rhythms correlate with cognition in Parkinson's disease. NPJ Parkinsons Dis 2021; 7:14. [PMID: 33589640 PMCID: PMC7884691 DOI: 10.1038/s41531-021-00158-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/22/2020] [Indexed: 01/30/2023] Open
Abstract
Patients with Parkinson's disease (PD) can have significant cognitive dysfunction; however, the mechanisms for these cognitive symptoms are unknown. Here, we used scalp electroencephalography (EEG) to investigate the cortical basis for PD-related cognitive impairments during interval timing, which requires participants to estimate temporal intervals of several seconds. Time estimation is an ideal task demand for investigating cognition in PD because it is simple, requires medial frontal cortical areas, and recruits basic executive processes such as working memory and attention. However, interval timing has never been systematically studied in PD patients with cognitive impairments. We report three main findings. First, 71 PD patients had increased temporal variability compared to 37 demographically matched controls, and this variability correlated with cognitive dysfunction as measured by the Montreal Cognitive Assessment (MOCA). Second, PD patients had attenuated ~4 Hz EEG oscillatory activity at midfrontal electrodes in response to the interval-onset cue, which was also predictive of MOCA. Finally, trial-by-trial linear mixed-effects modeling demonstrated that cue-triggered ~4 Hz power predicted subsequent temporal estimates as a function of PD and MOCA. Our data suggest that impaired cue-evoked midfrontal ~4 Hz activity predicts increased timing variability that is indicative of cognitive dysfunction in PD. These findings link PD-related cognitive dysfunction with cortical mechanisms of cognitive control, which could advance novel biomarkers and neuromodulation for PD.
Collapse
|
137
|
Mahmoudzadeh M, Wallois F, Tir M, Krystkowiak P, Lefranc M. Cortical hemodynamic mapping of subthalamic nucleus deep brain stimulation in Parkinsonian patients, using high-density functional near-infrared spectroscopy. PLoS One 2021; 16:e0245188. [PMID: 33493171 PMCID: PMC7833160 DOI: 10.1371/journal.pone.0245188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 12/23/2020] [Indexed: 12/02/2022] Open
Abstract
Subthalamic nucleus deep brain stimulation (STN-DBS) is an effective treatment for idiopathic Parkinson's disease. Despite recent progress, the mechanisms responsible for the technique's effectiveness have yet to be fully elucidated. The purpose of the present study was to gain new insights into the interactions between STN-DBS and cortical network activity. We therefore combined high-resolution functional near-infrared spectroscopy with low-resolution electroencephalography in seven Parkinsonian patients on STN-DBS, and measured cortical haemodynamic changes at rest and during hand movement in the presence and absence of stimulation (the ON-stim and OFF-stim conditions, respectively) in the off-drug condition. The relative changes in oxyhaemoglobin [HbO], deoxyhaemoglobin [HbR], and total haemoglobin [HbT] levels were analyzed continuously. At rest, the [HbO], [HbR], and [HbT] over the bilateral sensorimotor (SM), premotor (PM) and dorsolateral prefrontal (DLPF) cortices decreased steadily throughout the duration of stimulation, relative to the OFF-stim condition. During hand movement in the OFF-stim condition, [HbO] increased and [HbR] decreased concomitantly over the contralateral SM cortex (as a result of neurovascular coupling), and [HbO], [HbR], and [HbT] increased concomitantly in the dorsolateral prefrontal cortex (DLPFC)-suggesting an increase in blood volume in this brain area. During hand movement with STN-DBS, the increase in [HbO] was over the contralateral SM and PM cortices was significantly lower than in the OFF-stim condition, as was the decrease in [HbO] and [HbT] in the DLPFC. Our results indicate that STN-DBS is associated with a reduction in blood volume over the SM, PM and DLPF cortices, regardless of whether or not the patient is performing a task. This particular effect on cortical networks might explain not only STN-DBS's clinical effectiveness but also some of the associated adverse effects.
Collapse
Affiliation(s)
| | | | - Mélissa Tir
- Neurosurgery Department, CHU Amiens-Picardie, Amiens, France
| | - Pierre Krystkowiak
- Neurology Department, CHU Amiens-Picardie, Amiens, France
- Laboratory of Functional Neurosciences, University of Picardie Jules Verne, Amiens, France
| | - Michel Lefranc
- Neurosurgery Department, CHU Amiens-Picardie, Amiens, France
| |
Collapse
|
138
|
Julien C, Hache G, Dulac M, Dubrou C, Castelnovo G, Giordana C, Azulay JP, Fluchère F. The clinical meaning of levodopa equivalent daily dose in Parkinson's disease. Fundam Clin Pharmacol 2021; 35:620-630. [PMID: 33458868 DOI: 10.1111/fcp.12646] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/14/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022]
Abstract
Levodopa (L-dopa) remains the basis of pharmacological treatment of Parkinson's disease (PD). However, L-dopa therapy is associated with the development of complications and presents major challenges in the long-term treatment. Thus, other medications may be suggested to delay and/or reduce the doses of L-dopa in order to prevent complications. The interpretation of treatment evolution reported in clinical trials on PD may be tricky, especially due to some variability in medications and dose regimens. Some authors have suggested a conversion factor to generate a total L-dopa equivalent daily dose (LEDD), calculated as a sum of each parkinsonian medication. Therefore, LEDD provides an artificial summary of the total daily medication a patient is receiving, and to date, there is no report focusing on the clinical interpretation of this parameter. Thus, based on a 3-year, multi-center retrospective study assessing the impact of second-line therapy initiation on LEDD in PD patients, the aim of our article was to discuss LEDD as a quantitative outcome to estimate the impact of second-line therapies on medication regimens; and in the second part of the discussion, to provide a narrative review of the clinical outcomes associated with LEDD in the literature.
Collapse
Affiliation(s)
- Charlotte Julien
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Guillaume Hache
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Morgane Dulac
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Cléa Dubrou
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Pharmacie, Marseille, France
| | - Giovanni Castelnovo
- Centre Hospitalier Universitaire de Nimes, Service de Neurologie, Nîmes, France
| | - Caroline Giordana
- Centre Hospitalier Universitaire de Nice, Hôpital Pasteur 2, Service de Neurologie, Nice, France
| | - Jean-Philippe Azulay
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Neurologie pathologies du mouvement, Centre expert et de coordination interrégional pour la maladie de Parkinson, Marseille, France
| | - Frédérique Fluchère
- Assistance Publique Hôpitaux de Marseille, Hôpital de la Timone, Service de Neurologie pathologies du mouvement, Centre expert et de coordination interrégional pour la maladie de Parkinson, Marseille, France
| |
Collapse
|
139
|
Serotonergic control of the glutamatergic neurons of the subthalamic nucleus. PROGRESS IN BRAIN RESEARCH 2021; 261:423-462. [PMID: 33785138 DOI: 10.1016/bs.pbr.2020.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The subthalamic nucleus (STN) houses a dense cluster of glutamatergic neurons that play a central role in the functional dynamics of the basal ganglia, a group of subcortical structures involved in the control of motor behaviors. Numerous anatomical, electrophysiological, neurochemical and behavioral studies have reported that serotonergic neurons from the midbrain raphe nuclei modulate the activity of STN neurons. Here, we describe this serotonergic innervation and the nature of the regulation exerted by serotonin (5-hydroxytryptamine, 5-HT) on STN neuron activity. This regulation can occur either directly within the STN or at distal sites, including other structures of the basal ganglia or cortex. The effect of 5-HT on STN neuronal activity involves several 5-HT receptor subtypes, including 5-HT1A, 5-HT1B, 5-HT2C and 5-HT4 receptors, which have garnered the highest attention on this topic. The multiple regulatory effects exerted by 5-HT are thought to be modified under pathological conditions, altering the activity of the STN, or due to the benefits and side effects of treatments used for Parkinson's disease, notably the dopamine precursor l-DOPA and high-frequency STN stimulation. Originally understood as a motor center, the STN is also associated with decision making and participates in mood regulation and cognitive performance, two domains of personality that are also regulated by 5-HT. The literature concerning the link between 5-HT and STN is already important, and the functional overlap is evident, but this link is still not entirely understood. The understanding of this link between 5-HT and STN should be increased due to the possible importance of this regulation in the control of fronto-STN loops and inherent motor and non-motor behaviors.
Collapse
|
140
|
Cavallieri F, Fraix V, Bove F, Mulas D, Tondelli M, Castrioto A, Krack P, Meoni S, Schmitt E, Lhommée E, Bichon A, Pélissier P, Chevrier E, Kistner A, Seigneuret E, Chabardès S, Moro E. Predictors of Long-Term Outcome of Subthalamic Stimulation in Parkinson Disease. Ann Neurol 2021; 89:587-597. [PMID: 33349939 DOI: 10.1002/ana.25994] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/02/2020] [Accepted: 12/13/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study was undertaken to identify preoperative predictive factors of long-term motor outcome in a large cohort of consecutive Parkinson disease (PD) patients with bilateral subthalamic nucleus deep brain stimulation (STN-DBS). METHODS All consecutive PD patients who underwent bilateral STN-DBS at the Grenoble University Hospital (France) from 1993 to 2015 were evaluated before surgery, at 1 year (short-term), and in the long term after surgery. All available demographic variables, neuroimaging data, and clinical characteristics were collected. Preoperative predictors of long-term motor outcome were investigated by performing survival and univariate/multivariate Cox regression analyses. Loss of motor benefit from stimulation in the long term was defined as a reduction of less than 25% in the Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) part III scores compared to the baseline off-medication scores. As a secondary objective, potential predictors of short-term motor outcome after STN-DBS were assessed by performing univariate and multivariate linear regression analyses. RESULTS In the long-term analyses (mean follow-up = 8.4 ± 6.26 years, median = 10 years, range = 1-17 years), 138 patients were included. Preoperative higher frontal score and off-medication MDS-UPDRS part III scores predicted a better long-term motor response to stimulation, whereas the presence of vascular changes on neuroimaging predicted a worse motor outcome. In 357 patients with available 1-year follow-up, preoperative levodopa response, tremor dominant phenotype, baseline frontal score, and off-medication MDS-UPDRS part III scores predicted the short-term motor outcome. INTERPRETATION Frontal lobe dysfunction, disease severity in the off-medication condition, and the presence of vascular changes on neuroimaging represent the main preoperative clinical predictors of long-term motor STN-DBS effects. ANN NEUROL 2021;89:587-597.
Collapse
Affiliation(s)
- Francesco Cavallieri
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Neurology Unit, Neuromotor and Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Valérie Fraix
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France
| | - Francesco Bove
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Delia Mulas
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Heart, Rome, Italy.,Neurology Unit, Mater Olbia Hospital, Olbia, Italy
| | - Manuela Tondelli
- Neurology Unit, University Hospital Policlinico, Department of Biomedical, Metabolic, and Neural Science, Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Castrioto
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France
| | - Paul Krack
- Department of Neurology, Center for Parkinson's Disease and Movement Disorders, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sara Meoni
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France
| | - Emmanuelle Schmitt
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France
| | - Eugénie Lhommée
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France
| | - Amélie Bichon
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France
| | - Pierre Pélissier
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France
| | - Eric Chevrier
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France
| | - Andrea Kistner
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France
| | - Eric Seigneuret
- Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France.,Division of Neurosurgery, Grenoble Alpes University Hospital Center, Grenoble, France
| | - Stephan Chabardès
- Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France.,Division of Neurosurgery, Grenoble Alpes University Hospital Center, Grenoble, France
| | - Elena Moro
- Movement Disorders Unit, University Hospital Center, Grenoble Alpes University, Grenoble, France.,Grenoble Institute of Neurosciences, UGA INSERM U1216, Grenoble, France
| |
Collapse
|
141
|
Frisaldi E, Zamfira DA, Benedetti F. The subthalamic nucleus and the placebo effect in Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:433-444. [PMID: 34225946 DOI: 10.1016/b978-0-12-820107-7.00027-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The study of the placebo effect, or response, is related to the investigation of the psychologic component of different therapeutic rituals. The high rate of placebo responses in Parkinson's disease clinical trials provided the impetus for investigating the underlying mechanisms. Ruling out spontaneous remission and regression to the mean through the appropriate experimental designs, genuine psychologic placebo effects have been identified, in which both patients' expectations of therapeutic benefit and learning processes are involved. Specifically, placebo effects are associated with dopamine release in the striatum and changes in neuronal activity in the subthalamic nucleus, substantia nigra pars reticulata, and motor thalamus in Parkinson's disease, as assessed through positron emission tomography and single-neuron recording during deep brain stimulation, respectively. Conversely, verbal suggestions of clinical worsening or drug dose reduction induce nocebo responses in Parkinson's disease, which have been detected at both behavioral and electrophysiologic level. Important implications and applications emerge from this new knowledge. These include better clinical trial designs, whereby patients' expectations should always be assessed, as well as better drug dosage in order to reduce drug intake.
Collapse
Affiliation(s)
- Elisa Frisaldi
- Department of Neuroscience, University of Turin Medical School, Turin, Italy
| | | | - Fabrizio Benedetti
- Department of Neuroscience, University of Turin Medical School, Turin, Italy; Medicine and Physiology of Hypoxia, Plateau Rosà, Switzerland
| |
Collapse
|
142
|
Davids J, Ashrafian H. AIM in Neurodegenerative Diseases: Parkinson and Alzheimer. Artif Intell Med 2021. [DOI: 10.1007/978-3-030-58080-3_190-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
143
|
Piacentino M, Beggio G, Rustemi O, Zambon G, Pilleri M, Raneri F. Pneumocephalus in subthalamic deep brain stimulation for Parkinson's disease: a comparison of two different surgical techniques considering factors conditioning brain shift and target precision. Acta Neurochir (Wien) 2021; 163:169-175. [PMID: 33174114 DOI: 10.1007/s00701-020-04635-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/26/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Precise placement of electrodes in deep brain stimulation (DBS) may be influenced by brain shift caused by cerebrospinal fluid leaking or air inflow. We compared accuracy and treatment outcomes between a standard technique and one aiming at reducing brain shift. METHODS We retrospectively reviewed 46 patients (92 targets) treated with bilateral subthalamic-DBS for Parkinson's disease. The patients were divided into two groups: group A surgery was performed in supine position with standard burr hole, dural opening, fibrin glue and gelfoam plugging. Group B patients were operated in a semi-sitting position with direct dural puncture to reduce CSF loss. We analysed target deviation on head CT performed immediately after surgery and at 1 month merged with preoperative MRI planning. We recorded pneumocephalus volume, brain atrophy and target correction by intraoperative neurophysiology (ION). RESULTS In group A, the mean pneumocephalus volume was 10.55 cm3, mean brain volume 1116 cm3, mean target deviation 1.09 mm and ION corrected 70% of targets. In group B, mean pneumocephalus was 7.60 cm3 (p = 0.3048), mean brain volume 1132 cm3 (p = 0.6526), mean target deviation 0.64 mm (p = 0.0074) and ION corrected 50% of targets (p = 0.4886). Most leads' deviations realigned to the planned target after pneumocephalus reabsorbtion suggesting a deviation caused by displacement of anatomical structures due to brain shift. Definitive lead position was always decided with ION. CONCLUSIONS The modified DBS technique significantly reduced errors of electrode placement, though such difference was clinically irrelevant. ION corrected a high amount of trajectories in both groups (70% vs 50%). The choice of either strategy is acceptable.
Collapse
Affiliation(s)
- Massimo Piacentino
- Department of Neurosurgery, AULSS 8 Berica Ospedale San Bortolo, Viale Rodolfi, 37 36100, Vicenza, Italy
| | - Giacomo Beggio
- Department of Neurosurgery, AULSS 8 Berica Ospedale San Bortolo, Viale Rodolfi, 37 36100, Vicenza, Italy
| | - Oriela Rustemi
- Department of Neurosurgery, AULSS 8 Berica Ospedale San Bortolo, Viale Rodolfi, 37 36100, Vicenza, Italy
| | - Giampaolo Zambon
- Department of Neurosurgery, AULSS 8 Berica Ospedale San Bortolo, Viale Rodolfi, 37 36100, Vicenza, Italy
| | - Manuela Pilleri
- Department of Neurology, Casa di Cura Villa Margherita Hospital, Via Costa Colonna, 6 36057, Arcugnano, Italy
| | - Fabio Raneri
- Department of Neurosurgery, AULSS 8 Berica Ospedale San Bortolo, Viale Rodolfi, 37 36100, Vicenza, Italy.
| |
Collapse
|
144
|
Choi EH, Nwakalor C, Brown NJ, Lee J, Oh MY, Yang IH. Therapeutic potential of neuromodulation for demyelinating diseases. Neural Regen Res 2021; 16:214-217. [PMID: 32859766 PMCID: PMC7896214 DOI: 10.4103/1673-5374.290876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neuromodulation represents a cutting edge class of both invasive and non-invasive therapeutic methods which alter the activity of neurons. Currently, several different techniques have been developed - or are currently being investigated – to treat a wide variety of neurological and neuropsychiatric disorders. Recently, in vivo and in vitro studies have revealed that neuromodulation can also induce myelination, meaning that it could hold potential as a therapy for various demyelinating diseases including multiple sclerosis and progressive multifocal leukencepalopathy. These findings come on the heels of a paradigm shift in the view of myelin’s role within the nervous system from a static structure to an active co-regulator of central nervous system plasticity and participant in neuron-mediated modulation. In the present review, we highlight several of the recent findings regarding the role of neural activity in altering myelination including several soluble and contact-dependent factors that seem to mediate neural activity-dependent myelination. We also highlight several considerations for neuromodulatory techniques, including the need for further research into spatiotemporal precision, dosage, and the safety and efficacy of transcranial focused ultrasound stimulation, an emerging neuromodulation technology. As the field of neuromodulation continues to evolve, it could potentially bring forth methods for the treatment of demyelinating diseases, and as such, further investigation into the mechanisms of neuron-dependent myelination as well as neuro-imaging modalities that can monitor myelination activity is warranted.
Collapse
Affiliation(s)
- Elliot H Choi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH; Department of Ophthalmology, Gavin Herbert Eye Institute, School of Medicine, University of California; Department of Neurological Surgery, University of California, Irvine, CA, USA
| | - Chioma Nwakalor
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Nolan J Brown
- Department of Neurological Surgery, University of California, Irvine, CA, USA
| | - Joonho Lee
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael Y Oh
- Department of Neurological Surgery, University of California, Irvine, CA, USA
| | - In Hong Yang
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC, USA
| |
Collapse
|
145
|
Ehlen F, Al-Fatly B, Kühn AA, Klostermann F. Impact of deep brain stimulation of the subthalamic nucleus on natural language in patients with Parkinson's disease. PLoS One 2020; 15:e0244148. [PMID: 33373418 PMCID: PMC7771859 DOI: 10.1371/journal.pone.0244148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 12/03/2020] [Indexed: 12/30/2022] Open
Abstract
Background In addition to the typical motor symptoms, a majority of patients suffering from Parkinson’s disease experience language impairments. Deep Brain Stimulation of the subthalamic nucleus robustly reduces motor dysfunction, but its impact on language skills remains ambiguous. Method To elucidate the impact of subthalamic deep brain stimulation on natural language production, we systematically analyzed language samples from fourteen individuals (three female / eleven male, average age 66.43 ± 7.53 years) with Parkinson’s disease in the active (ON) versus inactive (OFF) stimulation condition. Significant ON-OFF differences were considered as stimulation effects. To localize their neuroanatomical origin within the subthalamic nucleus, they were correlated with the volume of tissue activated by therapeutic stimulation. Results Word and clause production speed increased significantly under active stimulation. These enhancements correlated with the volume of tissue activated within the associative part of the subthalamic nucleus, but not with that within the dorsolateral motor part, which again correlated with motor improvement. Language error rates were lower in the ON vs. OFF condition, but did not correlate with electrode localization. No significant changes in further semantic or syntactic language features were detected in the current study. Conclusion The findings point towards a facilitation of executive language functions occurring rather independently from motor improvement. Given the presumed origin of this stimulation effect within the associative part of the subthalamic nucleus, this could be due to co-stimulation of the prefrontal-subthalamic circuit.
Collapse
Affiliation(s)
- Felicitas Ehlen
- Department of Neurology, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Jüdisches Krankenhaus Berlin, Berlin, Germany
- * E-mail:
| | - Bassam Al-Fatly
- Department of Neurology, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea A. Kühn
- Department of Neurology, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Berlin, Germany
| | - Fabian Klostermann
- Department of Neurology, Humboldt-Universität zu Berlin and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
146
|
Martínez-Fernández R, Máñez-Miró JU, Rodríguez-Rojas R, Del Álamo M, Shah BB, Hernández-Fernández F, Pineda-Pardo JA, Monje MHG, Fernández-Rodríguez B, Sperling SA, Mata-Marín D, Guida P, Alonso-Frech F, Obeso I, Gasca-Salas C, Vela-Desojo L, Elias WJ, Obeso JA. Randomized Trial of Focused Ultrasound Subthalamotomy for Parkinson's Disease. N Engl J Med 2020; 383:2501-2513. [PMID: 33369354 DOI: 10.1056/nejmoa2016311] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The subthalamic nucleus is the preferred neurosurgical target for deep-brain stimulation to treat cardinal motor features of Parkinson's disease. Focused ultrasound is an imaging-guided method for creating therapeutic lesions in deep-brain structures, including the subthalamic nucleus. METHODS We randomly assigned, in a 2:1 ratio, patients with markedly asymmetric Parkinson's disease who had motor signs not fully controlled by medication or who were ineligible for deep-brain stimulation surgery to undergo focused ultrasound subthalamotomy on the side opposite their main motor signs or a sham procedure. The primary efficacy outcome was the between-group difference in the change from baseline to 4 months in the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) motor score (i.e., part III) for the more affected body side (range, 0 to 44, with higher scores indicating worse parkinsonism) in the off-medication state. The primary safety outcome (procedure-related complications) was assessed at 4 months. RESULTS Among 40 enrolled patients, 27 were assigned to focused ultrasound subthalamotomy (active treatment) and 13 to the sham procedure (control). The mean MDS-UPDRS III score for the more affected side decreased from 19.9 at baseline to 9.9 at 4 months in the active-treatment group (least-squares mean difference, 9.8 points; 95% confidence interval [CI], 8.6 to 11.1) and from 18.7 to 17.1 in the control group (least-squares mean difference, 1.7 points; 95% CI, 0.0 to 3.5); the between-group difference was 8.1 points (95% CI, 6.0 to 10.3; P<0.001). Adverse events in the active-treatment group were dyskinesia in the off-medication state in 6 patients and in the on-medication state in 6, which persisted in 3 and 1, respectively, at 4 months; weakness on the treated side in 5 patients, which persisted in 2 at 4 months; speech disturbance in 15 patients, which persisted in 3 at 4 months; facial weakness in 3 patients, which persisted in 1 at 4 months; and gait disturbance in 13 patients, which persisted in 2 at 4 months. In 6 patients in the active-treatment group, some of these deficits were present at 12 months. CONCLUSIONS Focused ultrasound subthalamotomy in one hemisphere improved motor features of Parkinson's disease in selected patients with asymmetric signs. Adverse events included speech and gait disturbances, weakness on the treated side, and dyskinesia. (Funded by Insightec and others; ClinicalTrials.gov number, NCT03454425.).
Collapse
Affiliation(s)
- Raúl Martínez-Fernández
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Jorge U Máñez-Miró
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Rafael Rodríguez-Rojas
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Marta Del Álamo
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Binit B Shah
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Frida Hernández-Fernández
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - José A Pineda-Pardo
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Mariana H G Monje
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Beatriz Fernández-Rodríguez
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Scott A Sperling
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - David Mata-Marín
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Pasqualina Guida
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Fernando Alonso-Frech
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Ignacio Obeso
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Carmen Gasca-Salas
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - Lydia Vela-Desojo
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - W Jeffrey Elias
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| | - José A Obeso
- From HM Centro Integral en Neurociencias AC (CINAC), University Hospital HM Puerta del Sur, CEU San Pablo University, Mostoles (R.M.-F., J.U.M.-M., R.R.-R., M.A., F.H.-F., J.A.P.-P., M.H.G.M., B.F.-R., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.), and the Network Center for Biomedical Research on Neurodegenerative Diseases, Carlos III Institute, Madrid (R.M.-F., R.R.-R., M.A., F.H.-F., J.A.P.-P., D.M.-M., P.G., F.A.-F., I.O., C.G.-S., L.V.-D., J.A.O.) - both in Spain; and the University of Virginia Health Sciences Center, Charlottesville (B.B.S., S.A.S., W.J.E.)
| |
Collapse
|
147
|
Zhang Q, Weber MA, Narayanan NS. Medial prefrontal cortex and the temporal control of action. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 158:421-441. [PMID: 33785154 DOI: 10.1016/bs.irn.2020.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Across species, the medial prefrontal cortex guides actions in time. This process can be studied using behavioral paradigms such as simple reaction-time and interval-timing tasks. Temporal control of action can be influenced by prefrontal neurotransmitters such as dopamine and acetylcholine and is highly relevant to human diseases such as Parkinson's disease, schizophrenia, and attention-deficit hyperactivity disorder (ADHD). We review evidence that across species, medial prefrontal lesions impair the temporal control of action. We then consider neurophysiological correlates in humans, primates, and rodents that might encode temporal processing and relate to cognitive-control mechanisms. These data have informed brain-stimulation studies in rodents and humans that can compensate for timing deficits. This line of work illuminates basic mechanisms of temporal control of action in the medial prefrontal cortex, which underlies a range of high-level cognitive processing and could contribute to new biomarkers and therapies for human brain diseases.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | - Matthew A Weber
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| | | |
Collapse
|
148
|
Guridi J, Gonzalez-Quarante LH. Revisiting Forel Field Surgery. World Neurosurg 2020; 147:11-22. [PMID: 33276174 DOI: 10.1016/j.wneu.2020.11.143] [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: 07/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Lesioning the Forel field or the subthalamic region is considered a possible treatment for tremoric patients with Parkinson disease, essential tremor, and other diseases. This surgical treatment was performed in the 1960s to 1970s and was an alternative to thalamotomy. Recently, there has been increasing interest in the reappraisal of stimulating and/or lesioning these targets, partly as a result of innovations in imaging and noninvasive ablative technologies, such as magnetic resonance-guided focused ultrasonography. OBJECTIVE We wanted to perform a thorough review of the subthalamic region, both from an anatomic and a surgical standpoint, to offer a comprehensive and updated analysis of the techniques and results reported for patients with tremor treated with different techniques. METHODS We performed a systematic review of the literature, gathering articles that included patients who underwent ablative or stimulation surgical techniques, targeting the pallidothalamic pathways (pallidothalamic tractotomy), cerebellothalamic pathway (cerebellothalamic tractotomy), or subthalamic area. RESULTS Pallidothalamic tractotomy consists of a reduced area that includes pallidofugal pathways. It may be considered an interesting target, given the benefit/risk ratio and the clinical effect, which, compared with pallidotomy, involves a lower risk of injury or involvement of vital structures such as the internal capsule or optic tract. Cerebellothalamic tractotomy and/or posterior subthalamic area are other alternative targets to thalamic stimulation or ablative surgery. CONCLUSIONS Based on the significant breakthrough that magnetic resonance-guided focused ultrasonography has meant in the neurosurgical world, some classic targets such as the pallidothalamic tract, Forel field, and posterior subthalamic area may be reconsidered as surgical alternatives for patients with movement disorders.
Collapse
Affiliation(s)
- Jorge Guridi
- Department of Neurosurgery, Clínica Universidad de Navarra, Navarra University, Pamplona, Spain
| | | |
Collapse
|
149
|
Engelhardt J, Caire F, Damon-Perrière N, Guehl D, Branchard O, Auzou N, Tison F, Meissner WG, Krim E, Bannier S, Bénard A, Sitta R, Fontaine D, Hoarau X, Burbaud P, Cuny E. A Phase 2 Randomized Trial of Asleep versus Awake Subthalamic Nucleus Deep Brain Stimulation for Parkinson's Disease. Stereotact Funct Neurosurg 2020; 99:230-240. [PMID: 33254172 DOI: 10.1159/000511424] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/07/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Asleep deep brain stimulation (DBS) for Parkinson's disease (PD) is being performed more frequently; however, motor outcomes and safety of asleep DBS have never been assessed in a prospective randomized trial. METHODS We conducted a prospective, randomized, noncomparative trial to assess the motor outcomes of asleep DBS. Leads were implanted in the subthalamic nucleus (STN) according to probabilistic stereotactic coordinates with a surgical robot under O-arm© imaging guidance under either general anesthesia without microelectrode recordings (MER) (20 patients, asleep group) or local anesthesia with MER and clinical testing (9 patients, awake group). RESULTS The mean motor improvement rates on the Unified Parkinson's Disease Rating Scale Part III (UPDRS-3) between OFF and ON stimulation without medication were 52.3% (95% CI: 45.4-59.2%) in the asleep group and 47.0% (95% CI: 23.8-70.2%) in the awake group, 6 months after surgery. Except for a subcutaneous hematoma, we did not observe any complications related to the surgery. Three patients (33%) in the awake group and 8 in the asleep group (40%) had at least one side effect potentially linked with neurostimulation. CONCLUSIONS Owing to its randomized design, our study supports the hypothesis that motor outcomes after asleep STN-DBS in PD may be noninferior to the standard awake procedure.
Collapse
Affiliation(s)
- Julien Engelhardt
- CHU de Bordeaux, Service de Neurochirurgie B, Bordeaux, France, .,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France, .,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France,
| | - François Caire
- Université de Limoges, CNRS, XLIM, UMR7252, Limoges, France.,CHU de Limoges, Service de Neurochirurgie, Limoges, France
| | - Nathalie Damon-Perrière
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CHU de Bordeaux, Service d'explorations Fonctionnelles du Système Nerveux, Bordeaux, France
| | - Dominique Guehl
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CHU de Bordeaux, Service d'explorations Fonctionnelles du Système Nerveux, Bordeaux, France
| | | | - Nicolas Auzou
- CHU de Bordeaux, Service de Neurologie, Bordeaux, France.,Laboratoire de Psychologie, Université de Bordeaux, Bordeaux, France
| | - François Tison
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CHU de Bordeaux, Service de Neurologie, Bordeaux, France
| | - Wassilios G Meissner
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CHU de Bordeaux, Service de Neurologie, Bordeaux, France
| | - Elsa Krim
- CH de Pau, Service de Neurologie, Pau, France
| | | | - Antoine Bénard
- CHU Bordeaux, Pôle de Santé Publique, Clinical Epidemiology Unit (USMR), Bordeaux, France
| | - Rémi Sitta
- CHU Bordeaux, Pôle de Santé Publique, Clinical Epidemiology Unit (USMR), Bordeaux, France
| | - Denys Fontaine
- CHU de Nice, Service de Neurochirurgie, Nice, France.,Université Côte d'Azur, Nice, France
| | - Xavier Hoarau
- Polyclinique de Navarre, Service de Neurochirurgie, Pau, France
| | - Pierre Burbaud
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CHU de Bordeaux, Service d'explorations Fonctionnelles du Système Nerveux, Bordeaux, France
| | - Emmanuel Cuny
- CHU de Bordeaux, Service de Neurochirurgie B, Bordeaux, France.,Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| |
Collapse
|
150
|
Isaacs BR, Mulder MJ, Groot JM, van Berendonk N, Lute N, Bazin PL, Forstmann BU, Alkemade A. 3 versus 7 Tesla magnetic resonance imaging for parcellations of subcortical brain structures in clinical settings. PLoS One 2020; 15:e0236208. [PMID: 33232325 PMCID: PMC7685480 DOI: 10.1371/journal.pone.0236208] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
7 Tesla (7T) magnetic resonance imaging holds great promise for improved visualization of the human brain for clinical purposes. To assess whether 7T is superior regarding localization procedures of small brain structures, we compared manual parcellations of the red nucleus, subthalamic nucleus, substantia nigra, globus pallidus interna and externa. These parcellations were created on a commonly used clinical anisotropic clinical 3T with an optimized isotropic (o)3T and standard 7T scan. The clinical 3T MRI scans did not allow delineation of an anatomically plausible structure due to its limited spatial resolution. o3T and 7T parcellations were directly compared. We found that 7T outperformed the o3T MRI as reflected by higher Dice scores, which were used as a measurement of interrater agreement for manual parcellations on quantitative susceptibility maps. This increase in agreement was associated with higher contrast to noise ratios for smaller structures, but not for the larger globus pallidus segments. Additionally, control-analyses were performed to account for potential biases in manual parcellations by assessing semi-automatic parcellations. These results showed a higher consistency for structure volumes for 7T compared to optimized 3T which illustrates the importance of the use of isotropic voxels for 3D visualization of the surgical target area. Together these results indicate that 7T outperforms c3T as well as o3T given the constraints of a clinical setting.
Collapse
Affiliation(s)
- Bethany R. Isaacs
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
- Department of Experimental Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martijn J. Mulder
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
- Psychology and Social Sciences, University of Utrecht, Utrecht, The Netherlands
| | - Josephine M. Groot
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
| | - Nikita van Berendonk
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
| | - Nicky Lute
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
- Clinical Neuropsychology, Vrije University, Amsterdam, The Netherlands
| | - Pierre-Louis Bazin
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
- Max Planck Institute for Human, Cognitive and Brain Sciences, Leipzig, Germany
| | - Birte U. Forstmann
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
| | - Anneke Alkemade
- University of Amsterdam, Integrative Model-Based Cognitive Neuroscience Research Unit, Amsterdam, The Netherlands
| |
Collapse
|